US7993627B2 - Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal - Google Patents

Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal Download PDF

Info

Publication number
US7993627B2
US7993627B2 US10/335,035 US33503502A US7993627B2 US 7993627 B2 US7993627 B2 US 7993627B2 US 33503502 A US33503502 A US 33503502A US 7993627 B2 US7993627 B2 US 7993627B2
Authority
US
United States
Prior art keywords
app
peptide
antibody
compound
amount
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US10/335,035
Other versions
US20030148392A1 (en
Inventor
Martin Citron
Dennis J. Selkoe
Peter A. Seubert
Dale B. Schenk
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Brigham and Womens Hospital Inc
Elan Pharmaceuticals LLC
Original Assignee
Brigham and Womens Hospital Inc
Elan Pharmaceuticals LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=24670990&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US7993627(B2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US08/079,511 external-priority patent/US5766846A/en
Application filed by Brigham and Womens Hospital Inc, Elan Pharmaceuticals LLC filed Critical Brigham and Womens Hospital Inc
Priority to US10/335,035 priority Critical patent/US7993627B2/en
Publication of US20030148392A1 publication Critical patent/US20030148392A1/en
Application granted granted Critical
Publication of US7993627B2 publication Critical patent/US7993627B2/en
Adjusted expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4709Amyloid plaque core protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer

Definitions

  • the present invention relates generally to neurology and, more particularly, to assays, such as immunoassays, for screening for compounds that specifically alter the production of various isoforms of A ⁇ .
  • AD Alzheimer's disease
  • AD is a degenerative brain disorder characterized clinically by progressive loss of memory, cognition, reasoning, judgment and emotional stability that gradually leads to profound mental deterioration and ultimately death.
  • AD is a very common cause of progressive mental failure (dementia) in aged humans and is believed to represent the fourth most common medical cause of death in the United States.
  • AD has been observed in all races and ethnic groups worldwide and presents a major present and future public health problem. The disease is currently estimated to affect about two to three million individuals in the United States alone. AD is at present incurable. No treatment that effectively prevents AD or reverses its symptoms or course is currently known.
  • senile plaques and neurofibrillary tangles The brains of individuals with AD exhibit characteristic lesions termed senile plaques and neurofibrillary tangles. Large numbers of these lesions are generally found in patients with AD in several areas of the human brain important for memory and cognitive function. Smaller numbers of these lesions in a more restricted anatomical distribution are sometimes found in the brains of aged humans who do not have clinical AD.
  • Senile plaques and vascular amyloid deposits also characterize the brains of individuals beyond a certain age with Trisomy 21 (Down's Syndrome) and Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type (HCHWA-D).
  • amyloid- ⁇ peptide A ⁇ was first purified and a partial amino acid sequence reported in Glenner and Wong (1984) Biochem. Biophys. Res. Commun. 120:885-890. The isolation procedure and the sequence data for the first 28 amino acids are described in U.S. Pat. No. 4,666,829. Forms of A ⁇ having amino acids beyond number 40 were first reported by Kang et al. (1987) Nature 325:733-736.
  • a ⁇ 42(43)-containing senile plaques are the major species of senile plaques in sporadic AD brains.
  • Iwatsubo et al. (1995) Annals of Neurology 37:294-299 and Lemere et al. (1996) Neurobiology of Disease 3:16-32 reported that A ⁇ 42(43) is the major constituent of senile plaques in Down's syndrome brains and is the initially deposited A ⁇ species in the development of AD-type neuropathological legions in these patients.
  • Gravina et al., (1995) J. Biol. Chem . 270:7013-7016 reported both biochemical and immunocytochemical evidence that A ⁇ 42(43) peptides were the most abundant constituents of senile plaques in AD brains and exceeded the amounts of A ⁇ 40 peptides in such plaques.
  • a ⁇ is a small fragment of a much larger precursor protein, referred to as the ⁇ -amyloid precursor protein (APP), that is normally produced by cells in many tissues of various animals, including humans.
  • APP ⁇ -amyloid precursor protein
  • Knowledge of the structure of the gene encoding APP has demonstrated that A ⁇ arises as a peptide fragment that is cleaved from the carboxy-terminal end of APP by as-yet-unknown enzymes (proteases).
  • proteases as-yet-unknown enzymes
  • the precise biochemical mechanism by which the A ⁇ fragment is cleaved from APP and subsequently deposited as amyloid plaques in the cerebral tissue and in the walls of cerebral and meningeal blood vessels is currently unknown.
  • a double mutation changing lysine 670 -methionine 671 to asparagine 670 -leucine 671 was reported in a Swedish family with familial AD in 1992 (Mullan et al. (1992) Nature Genet 1:345-347) and is referred to as the Swedish APP variant.
  • a ⁇ (42) is the major plaque component (Kang et al. (1987) Nature 325:733-736; Iwatsubo et al. (1994) Neuron 13:45-53; Iwatsubo et al. (1995) Ann. Neurol . 37:294-299; Gravina et al. (1995) J. Biol. Chem . 270:7013-7016; Lemere et al. (1996) Neurobiology of Disease 3:16-32).
  • proteases involved in the major steps of APP processing have been definitively identified, including ⁇ -secretase, the protease which generates the C-terminus of A ⁇ . It has generally been assumed that the same protease(s) generate both A ⁇ (40) and A(42) and it has been shown that both forms share a common secretory mechanism which involves acidic intracellular compartments such as the late Golgi or early endosomes (Koo and Squazzo (1994) J. Biol. Chem . 269:17386-17389; Asami-Odaka et al. (1995) Biochemistry 34:10272-10278). Recently, Higaki et al.
  • AD Alzheimer's disease
  • APP processing of APP is believed to involve several specific cleavages by proteases.
  • the enzyme that cleaves APP between amino acids 671/672 (referring to the ⁇ APP 770 isoform) is called ⁇ -secretase.
  • the enzyme that cleaves between amino acids 687/688 of APP (16/17 of A ⁇ ) is called ⁇ -secretase.
  • ⁇ -secretase The enzyme that cleaves between amino acids 687/688 of APP (16/17 of A ⁇ ) is called ⁇ -secretase.
  • a single enzyme called ⁇ -secretase.
  • a compound can inhibit the production of A ⁇ (40) but not A ⁇ (42).
  • compounds, thought to inhibit the production A ⁇ in general actually inhibit production of A ⁇ (40) but not A ⁇ (42). This indicates that multiple ⁇ -secretase mechanisms are at work which can be pharmacologically dissociated.
  • a ⁇ (42) is the major component of ⁇ -amyloid plaques and initiates amyloid plaque formation in AD patients, it is important to have tools to screen compounds to identify those that specifically inhibit the production of A ⁇ (42) and A ⁇ (40), either simultaneously or separately.
  • the current invention provides such assays.
  • This invention provides methods for determining whether a compound alters the amount of at least one A ⁇ (x- ⁇ 41) peptide produced by a cell.
  • the methods involve administering the compound to a culture comprising the cell; measuring the amount of the A ⁇ (x- ⁇ 41) peptide, specifically, in a sample from the culture; and determining whether the measured amount is different than the amount expected in a sample from a culture comprising the cell to which no compound has been administered. A difference between the measured amount and the expected amount indicates that the compound alters the amount of an A ⁇ (x- ⁇ 41) peptide produced by the cell.
  • this invention provides methods for determining whether a compound alters the amount of at least one A ⁇ (x- ⁇ 41) peptide produced by a cell and alters the amount of either total A ⁇ or at least one A ⁇ (x- ⁇ 40) peptide produced by the cell.
  • the methods involve administering the compound to a culture comprising the cell; measuring the amount of the A ⁇ (x- ⁇ 41) peptide, specifically, in a sample from the culture; measuring the amount of total A ⁇ or the A ⁇ (x- ⁇ 40) peptide, specifically, in a sample from the culture; and determining whether the measured amounts are different than the amounts expected in a sample from a culture comprising the cell to which no compound has been administered. Differences between the measured amounts and the expected amounts indicate that the compound alters the amount of the A ⁇ (x- ⁇ 41) peptide by a cell and/or the amount of total A ⁇ or the A ⁇ (x- ⁇ 40) peptide by the cell.
  • the amount of the A ⁇ peptides are measured by immunoassay and, in particular, sandwich immunoassay comprising capture binding substances bound to a solid phase and a labeled detection binding substance.
  • the capture antibody preferably is specific for A ⁇ (x- ⁇ 41) peptides, e.g., raised against peptide NH 2 -Cys-NH—CH 2 —(CH 2 ) 5 —CO-GLMVGGVVIA-COOH (SEQ ID NO: 4).
  • the detection binding substance in this assay can be an antibody specific for A ⁇ peptides whose amino-terminal amino acid is no. 1 of A ⁇ , or can be specific for an epitope within the junction region of A ⁇ .
  • the capture binding substance for measuring the amount of at least one of A ⁇ (x- ⁇ 41) peptide is specific for an epitope within the junction region of A ⁇ and the detection binding substance is an antibody specific for A ⁇ (x- ⁇ 41).
  • the capture binding substance preferably is an antibody specific for A ⁇ (x- ⁇ 40) peptides, e.g., raised against the peptide NH 2 -Cys-NH—CH 2 —(CH 2 ) 5 —CO-GLMVGGVV-COOH (SEQ ID NO: 5).
  • the labeled detection binding substance can be an antibody specific for the A ⁇ peptides whose amino-terminal amino acid is no. 1 of A ⁇ or an antibody specific for an epitope within the junction region of A ⁇ .
  • the capture binding substance for measuring the amount of at least one of A ⁇ (x- ⁇ 40) peptide is specific for an epitope within the junction region of A ⁇ and the detection binding substance is an antibody specific for A ⁇ (x- ⁇ 40).
  • the capture binding substance preferably is an antibody specific for an epitope within the junction region of A ⁇ .
  • the detection binding substance preferably is specific for A ⁇ peptides whose amino-terminal amino acid is no. 1 of A ⁇ .
  • the step of measuring the amount of the A ⁇ (x- ⁇ 41) peptide, total A ⁇ or the A ⁇ (x- ⁇ 40) peptide in a sample from the culture comprises: pulsing the culture with a radioactive label for protein; chasing the culture without a radioactive label; administering the compound to the cell during the chase period; contacting a sample from the culture with a binding substance specific for A ⁇ (x- ⁇ 41) peptides; contacting a sample from the culture with a binding substance specific for total A ⁇ or A ⁇ (x- ⁇ 40) peptide; and determining the amount of radioactive label attached to the binding substances.
  • the culture comprises primary human neurons, primary neurons from a transgenic PDAPP mouse (i.e., a transgenic mouse whose cells harbor a PDAPP construct), a 293 human kidney cell line, a human neuroglioma cell line, a human HeLa cell line, a primary endothelial cell line, a primary human fibroblast line, a primary lymphoblast line, human mixed brain cells, or a Chinese hamster ovary (CHO) cell line.
  • a transgenic PDAPP mouse i.e., a transgenic mouse whose cells harbor a PDAPP construct
  • a 293 human kidney cell line a human neuroglioma cell line
  • a human HeLa cell line a primary endothelial cell line
  • a primary human fibroblast line a primary lymphoblast line
  • human mixed brain cells or a Chinese hamster ovary (CHO) cell line.
  • the cell is a host cell transfected with a recombinant expression vector encoding a human APP, e.g., a Hardy mutation such as V717F or the Swedish mutant; causing the cell to overproduce A ⁇ (x- ⁇ 41) peptides.
  • the methods further comprise the step of determining whether the compound is toxic to the cell.
  • kits for specifically detecting at least one A ⁇ (x- ⁇ 41) peptide and at least one A ⁇ (x- ⁇ 40) peptide in a sample include a binding substance specific for A ⁇ (x- ⁇ 41) peptides; and a binding substance specific for A ⁇ (x- ⁇ 40) peptides.
  • kits for specifically detecting at least one A ⁇ (x- ⁇ 41) peptide and either total A ⁇ or at least one A ⁇ (x- ⁇ 40) peptide in a sample in a sandwich immunoassay include at least two different binding substances for measuring the amount of A ⁇ (x- ⁇ 41) peptide; and at least two different binding substances for measuring the amount of total A ⁇ or A ⁇ (x- ⁇ 40) peptides.
  • this invention provides methods for determining whether a compound alters the amount of at least one A ⁇ (x- ⁇ 41) peptide produced by a non-human mammal and alters the amount of either total A ⁇ or at least one A ⁇ (x- ⁇ 40) peptide produced in the non-human mammal.
  • the methods involve measuring a first amount of the A ⁇ (x- ⁇ 41) peptide in a sample from a non-human animal used as a model of Alzheimer's disease; measuring a first amount of total A ⁇ or the A ⁇ (x- ⁇ 40) peptide in a sample from the non-human animal; administering the compound to the non-human animal; measuring a second amount of the A ⁇ (x- ⁇ 41) peptide in a sample from the non-human animal; measuring a second amount of total A ⁇ or the A ⁇ (x- ⁇ 40) peptide in a sample from the non-human animal; and comparing the first amounts with the second amounts.
  • the non-human animal is a rodent, in particular, a mouse.
  • the non-human animal can harbor a copy of an expressible transgene sequence which encodes a Hardy mutation (e.g., V717F) or the Swedish mutation of human ⁇ -amyloid precursor protein (APP).
  • a Hardy mutation e.g., V717F
  • APP human ⁇ -amyloid precursor protein
  • FIG. 1 Shows standard curves of A ⁇ (1-40) and A ⁇ (1-42) with 2G3 as a capture antibody.
  • FIG. 2 Shows standard curves of A ⁇ (1-40) and A ⁇ (1-42) with 21F12 as a capture antibody.
  • FIG. 3 Shows that compound MDL 28170 influences ⁇ APP metabolism. (0) untreated K695sw cells; (200) K695sw cells treated with 200 ⁇ M MDL 28170 during the chase period. Lanes 1,2: aliquot of total chase media run directly on the gel. Lanes 3,4: antibody 1736 immunoprecipitation of chase media. Lanes 5,6: antibody 192sw immunoprecipitation of chase media. Note that ⁇ -cut APPs (lanes 5,6) runs slightly below ⁇ -cut APPs (lanes 3,4), as expected. Lanes 7,8: antibody C7 immunoprecipitation of cell lysates. Lanes 9,10: antibody 1282 immunoprecipitation of cell lysates.
  • FIGS. 4A-4B Differential inhibition of A ⁇ (42) and A ⁇ (40) formation.
  • A Labeled K695sw cells were chased with the indicated concentrations of MDL 28170, and the chase media were precipitated with 21F12 (upper panel) followed by antibody 1282 (lower panel).
  • B Quantitation of the effect of 200 ⁇ M MDL 28170 on A ⁇ and p3 in the chase media by phosphorimaging. The bars show the relative pixel number compared to an untreated control (set at 100%).
  • FIGS. 5A-5F Differential inhibition of A ⁇ production by K695sw cells under a variety of conditions.
  • A treatment with 1 ⁇ M PDBu
  • P decreases both A ⁇ (42) and A ⁇ total .
  • Upper panel antibody 21F12 precipitation
  • lower panel subsequent antibody 1282 precipitation.
  • (B-E) K695sw cells were chased with (M) or without (0) 200 ⁇ M MDL 28170, and the chase media were immunoprecipitated (B) first with antibody BC05 (upper panel) and then with antibody 1282 (lower panel); (C) first with antibody C42 (upper panel) and then with antibody 1282 (lower panel); (D) first with antibody 2G3 (upper panel) and then with antibody 21F12 (lower panel); (E) after splitting the medium into two aliquots, either with antibody 21F12 (upper panel) or antibody 1282 (lower panel). (F) cells were labeled for 3 h in the presence of 100 ⁇ M MDL 28170, and the medium was immunoprecipitated first with antibody 21F12 (upper panel) and then with antibody 1282 (lower panel).
  • FIGS. 6A-6C Differential inhibition of A ⁇ production in different cell types. 35S-methionine labeled cells were chased with the indicated concentrations of MDL 28170 and precipitated with antibody 21F12 (upper panel) followed by antibody 1282 (lower panel).
  • A V717F cells. The relatively low APP expression leads to a faint A ⁇ 42 band.
  • B CHO695 cells. Note that in CHO cells the p3 brands migrate as doublets, as described (Koo and Squazzo, (1994) “Evidence that production and release of amyloid ⁇ -protein involves the endocytic pathway.” J. Biol. Chem . 269:17386-17389).
  • C SKN695 cells. While A ⁇ (42) and p3(42) are slightly increased at 200 ⁇ M MDL 28170, A ⁇ total and p3 total are decreased.
  • FIG. 7 Shows a summary sketch of some of the known proteolytic processing pathways of APP.
  • FIG. 8A Shows a schematic of a combination cDNA/genomic coding sequence allowing alternative splicing of the KPI and OX-2 exons.
  • FIG. 8B Shows a schematic of a combination cDNA/genomic coding sequence bearing, a mutation at position 717 and allowing alternative splicing of the KPI and OX-2 exons.
  • FIG. 9 Diagram of the intermediate constructs used to construct the APP splicing cassette and the PDAPP vector.
  • FIGS. 10A-H The DNA sequence (SEQ ID NO: 2) and the deduced amino acid sequence (SEQ ID NO: 3) of APP770.
  • FIG. 11 Diagram, of the genomic region of APP present in the PDAPP construct. The sizes of original introns 6, 7 and 8, as well as the sizes of the final introns are indicated in the diagram. The locations of the deletions in introns 6 and 8 present in the PDAPP construct also are indicated.
  • FIG. 12 Schematic map of the PDAPP vector, a combination cDNA/genomic APP construct.
  • FIGS. 13A-13O depict the percent inhibition of A ⁇ by several compounds using the brain cell culture method described in Example VIII.
  • the charts show inhibition of total A ⁇ (“% A ⁇ inhibition”), inhibition of A ⁇ (42) (“% 1-42 inhibition”) and inhibition of the metabolism of MTT (“% MTT inhibition”) (greater inhibition indicates greater cytotoxicity).
  • binding substance refers to a polypeptide substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof, which specifically bind and recognize an analyte (antigen).
  • the recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as the myriad immunoglobulin variable region genes.
  • Antibodies exist, e.g., as intact immunoglobulins or as a number of well characterized fragments produced by digestion with various peptidases. This includes, e.g., Fab′ and F(ab)′ 2 fragments.
  • binding substance also includes antibody fragments either produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA methodologies.
  • immunoassay is an assay that utilizes a binding substance to specifically bind an analyte.
  • the immunoassay is characterized by the use of specific binding properties of a particular antibody to isolate, target, and/or quantify the analyte.
  • the specified binding substances bind preferentially to a particular protein and do not bind in a significant amount to other proteins present in the sample.
  • Specific binding to a protein under such conditions requires an antibody that is selected for its specificity for a particular protein.
  • a variety of immunoassay formats may be used to select binding substances specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
  • a “label” is a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means.
  • useful labels include 32 P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, dioxigenin, or haptens and proteins for which antisera or monoclonal antibodies are available.
  • Binding substances can be made detectible, e.g., by incorporating a radio-label into the peptide, and used to detect antibodies specifically reactive with the peptide.
  • a label often generates a measurable signal, such as radioactivity, fluorescent light or enzyme activity, which can be used to quantitate the amount of bound label.
  • Alzheimer's disease is characterized by the initial deposition of A ⁇ (x- ⁇ 41) in the form of amyloid plaques in the brain. Therefore, effective treatments for AD are expected to decrease the production of these peptides, whereas agents that hasten progress of the disease are expected to increase production of the peptide.
  • Prior screening methods looked for compounds that decreased total A ⁇ . However, since A ⁇ (x- ⁇ 41) peptides are a small fraction of total A ⁇ , those assays could not determine whether the compound specifically inhibited A ⁇ (x- ⁇ 41) peptides. As results described herein indicate, compounds can alter the production of A ⁇ (x- ⁇ 40) but not A ⁇ (x- ⁇ 41).
  • a ⁇ (42) is the major constituent of neuritic plaques, it is useful to identify compounds that specifically inhibit the production of A ⁇ (x- ⁇ 41) peptides, either in addition to, or instead of A ⁇ (x- ⁇ 40) peptides. Accordingly, this invention provides methods for screening compounds that specifically elevate or decrease the production of the amount of A ⁇ (x- ⁇ 41) by a cell and compounds that elevate or decrease production of both A ⁇ (x- ⁇ 41) and A ⁇ (x- ⁇ 40) (e.g., total A ⁇ ), or of one or the other of these peptides.
  • Compounds that decrease production of A ⁇ (x- ⁇ 41) are candidates for use in treating the disease, while compounds that increase its production may hasten the disease and are to be avoided by humans.
  • Screening methods of this invention for determining whether a test compound specifically alters the amount of A ⁇ (x- ⁇ 41) produced by a cell involve administering the compound to the cell, usually in culture, measuring the amount of A ⁇ (x- ⁇ 41) specifically produced by the cell, and determining whether this amount is greater than, less than or the same as an amount the cell is expected to produce in the absence of the compound. If the amounts are different, then the compound affects the production of A ⁇ (x- ⁇ 41) by the cell. This amount can be measured, for example, in a sample from the culture, such as medium conditioned by the cell in culture, or in extracts derived from cells harvested from the culture.
  • the expected amount generally will be a control amount determined by measuring A ⁇ (x- ⁇ 41) produced by the cell in the absence of the compound. However, one also may determine the expected amount by extrapolation; measuring the amount of A ⁇ (x- ⁇ 41) produced upon administration of different amounts of the compound to the cell, and using these figures to calculate the expected amount. In certain instances measuring a control amount for the purposes of comparison may not be necessary because the effect of the compound on A ⁇ (x- ⁇ 41) production is clear. For example, a compound may render A ⁇ (x- ⁇ 41) undetectable in a cell that normally produces detectable amounts, indicating that the compound decreases A ⁇ (x- ⁇ 41) production from the amount expected in its absence.
  • this invention provides screening methods for determining whether a compound alters the production of A ⁇ (x- ⁇ 41) by a cell to a different degree than it alters the production of total A ⁇ or A ⁇ (x- ⁇ 40) by the cell. These methods are useful for determining whether a compound alters the production of A ⁇ (x- ⁇ 41) in addition to total A ⁇ , or alters the production of one or the other of A ⁇ (x- ⁇ 41) and A ⁇ (x- ⁇ 40). The methods involve administering the compound to the cell (usually in culture). Then, the degree to which the compound alters the production of A ⁇ (x- ⁇ 41), specifically, by the cell is determined.
  • the degree to which the compound alters the production of total A ⁇ or A ⁇ (x- ⁇ 40) by the cell also is determined. Then, the degrees are compared. The comparison indicates whether the compound alters the production of A ⁇ (x- ⁇ 41) instead of or in addition to A ⁇ (x- ⁇ 40).
  • Determining the degree to which a compound alters the production of one or the other peptide generally involves measuring the specific amount of the peptide in a sample from the culture; and comparing it with the amounts expected in a sample from a culture comprising the cell to which no compound has been administered.
  • amyloid- ⁇ peptide As used herein refer to an approximately 4.2 kD protein which, in the brains of AD, Down's Syndrome, HCHWA-D and some normal aged subjects, forms the subunit of the amyloid filaments comprising the senile (amyloid) plaques and the amyloid deposits in small cerebral and meningeal blood vessels (amyloid angiopathy).
  • a ⁇ can occur in a filamentous polymeric form (in this form, it exhibits the Congo-red and thioflavin-S dye-binding characteristics of amyloid described in connection therewith).
  • a ⁇ can also occur in a non-filamentous form (“preamyloid” or “amorphous” or “diffuse” deposits) in brain tissue, in which form no birefringent staining by Congo red occurs.
  • preamyloid or “amorphous” or “diffuse” deposits
  • Congo red form no birefringent staining by Congo red occurs.
  • a ⁇ is an approximately 39-43 amino acid fragment of a large membrane-spanning glycoprotein, referred to as the ⁇ -amyloid precursor protein (APP), encoded by a gene on the long arm of human chromosome 21. Forms of A ⁇ longer than 43 amino acids are also contemplated herein.
  • a ⁇ is further characterized by its relative mobility in SDS-polyacrylamide gel electrophoresis or in high performance liquid chromatography (HPLC).
  • HPLC high performance liquid chromatography
  • a ⁇ also refers to related polymorphic forms of A ⁇ , including those that result from mutations in the A ⁇ region of the APP gene.
  • a ⁇ fragment refers to fragments and degradation products of A ⁇ which are generated at low concentrations by mammalian cells. Particular A ⁇ fragments have a molecular weight of approximately 3 kD and are presently believed to include peptides with, for example, amino acid residues 3-34, 6-27, 6-34, 6-35, 6-42, 11-34, 11-40, 11-43, 12-43, 17-40 and 17-42 of A ⁇ (Vigo-Pelfrey et al. (1993) J. Neurochem . 61:1965-1968).
  • a ⁇ (x- ⁇ 41) refers to A ⁇ or an A ⁇ fragment whose amino-terminus begins at amino acid number 1 of A ⁇ or thereafter (i.e., which is amino-terminally truncated), and whose carboxy-terminus extends beyond amino acid number 40.
  • These peptides and fragments comprise a heterogenous group. For example, A ⁇ (6-42), A ⁇ (11-43) and A ⁇ (12-43) all have been found in the CSF. However, this list is not meant to be exclusive. Other peptides from among the group are presumed to exist in the culture media of cells expressing APP and are detectable with the methods described herein.
  • a ⁇ (42) refers to A ⁇ or an A ⁇ fragment whose C-terminal amino acid is # 42 of A ⁇ .
  • a ⁇ (x- ⁇ 40) refers to A ⁇ or an A ⁇ fragment whose amino-terminus begins at amino acid number 1 of A ⁇ or which is amino-terminally truncated, and whose carboxy-terminus extends no further than amino acid number 40. These peptides and fragments also comprise a heterogenous group.
  • a ⁇ (40) refers to A ⁇ or an A ⁇ fragment whose C-terminal amino acid is #40 of A ⁇ .
  • p3 refers to a peptide whose amino acid sequence is substantially similar to A ⁇ , but whose amino-terminal amino acid begins at amino acid 17 of A ⁇ .
  • p3 fragment refers to fragments and degradation products of p3. Whereas p3 is produced through a different processing pathway than A ⁇ , for the purposes of the detection methods of this invention, p3 and p3 fragments are considered to be a subset of A ⁇ peptides, because certain detection techniques that recognize A ⁇ solely from the carboxy-terminus generally also will recognize p3. Also it is shown that the same apparent mechanisms generate the p3 and A ⁇ carboxy-termini.
  • a ⁇ junction region refers to a region of A ⁇ which is centered at the site between amino acid residues 16 and 17 (Lys 16 and Leu 17 ), which is a principal target for proteolytic processing of APP.
  • processing referred to as “ ⁇ -secretory” processing, results in a variety of APP fragments which may, for example, terminate at amino acid 16 of A ⁇ and which, therefore, are potentially immunologically cross-reactive with antibodies to the intact A ⁇ molecule which are to be used in the methods of the present invention.
  • Antibodies raised against a synthetic peptide including amino acid residues 13-28 have been found to display the requisite specificity for the junction region.
  • APP amyloid- ⁇ precursor protein
  • APP is defined as a polypeptide that is encoded by a gene of the same name localized in humans on the long arm of chromosome 21 and that includes the A ⁇ region within the carboxyl third of its coding region.
  • APP is a glycosylated, single-membrane-spanning protein expressed in a wide variety of cells in many mammalian tissues. Examples of specific isotypes of APP which are currently known to exist in humans are the 695-amino acid polypeptide described by Kang et al. (1987) Nature 325:733-736; the 751-amino acid polypeptide described by Ponte et al.
  • Alzheimer's disease which includes familial Alzheimer's disease
  • Down's Syndrome Down's Syndrome
  • HCHWA-D Advanced aging of the brain
  • test cells used in the methods of this invention generally are ones that are able to secrete A ⁇ (x- ⁇ 41).
  • In vitro monitoring of A ⁇ (x- ⁇ 41) levels in conditioned medium from a suitable cell culture may be used for drug screening. By growing cells under conditions which result in the secretion of A ⁇ (x- ⁇ 41) into the culture medium, and exposing the cells to test compounds, the effect of these test compounds on A ⁇ (x- ⁇ 41) secretion can be observed.
  • Suitable cell lines include human and animal cell lines, such as, preferably, primary human neurons, and primary neurons from transgenic mice harboring human APP genes, e.g., cells from a transgenic PDAPP animal (e.g., mouse), as well as a 293 human kidney cell line, a human neuroglioma cell line, a human HeLa cell line, a primary endothelial cell line (e.g., HUVEC cells), a primary human fibroblast line or a primary lymphoblast line (including endogenous cells derived from patients with APP mutations), a primary human mixed brain cell culture (including neurons, astrocytes and neuroglia), or a Chinese hamster ovary (CHO) cell line.
  • human and animal cell lines such as, preferably, primary human neurons, and primary neurons from transgenic mice harboring human APP genes, e.g., cells from a transgenic PDAPP animal (e.g., mouse), as well as a 293 human kidney cell line, a human neurogliom
  • V717I valine to isoleucine at position 717 in the APP770 numbering system.
  • Cell lines which preferentially increase the levels or ratios of A ⁇ (x- ⁇ 41) would be particularly useful in the methods of invention.
  • Useful mutants at position 717 include V717F, V717I or V717G.
  • APP variants which overproduce A ⁇ .
  • overproduce it is meant that the amount of A ⁇ produced from the variant APP will be at least about one-and-a-half times and preferably at least two or five times greater than the amount produced from any or all of the normal APP isoforms, e.g., the 695, 751, and 770 amino acid isoforms which have been previously described.
  • Particularly preferred are APP variants having one or several amino acid substitutions directly amino-terminal of the A ⁇ cleavage site.
  • K293 cells which express an APP 695 DNA bearing a double mutation found in a Swedish FAD family produce approximately five-to-eight-fold more A ⁇ than cells expressing normal APP (Citron et al. (1992) Nature , 360:672-674).
  • the mutation at residue 596 appears to be principally responsible for the increase.
  • Host cells transfected with a recombinant expression vector that encodes APP also are useful as cells in the screening methods of this invention.
  • a plasmid that carries sequences encoding APP is pCMV695 (Selkoe et al. (1988) Proc. Natl. Acad. Sci USA 85:7341-7345).
  • Nucleic acids encoding APP can be obtained by methods known in the art.
  • a nucleic acid encoding an APP can be isolated by polymerase chain reaction of cDNA or genomic DNA from a human brain cDNA library or a human genomic library using primers based on the DNA sequence of APP.
  • PCR methods are described in, for example, U.S. Pat. No. 4,683,195; Mullis et al. (1987) Cold Spring Harbor Symp. Quant. Biol . 51:263; and Erlich, ed., PCR Technology , (Stockton Press, NY, 1989).
  • Mutant versions of APP can be made by site-specific mutagenesis of other nucleic acids encoding APP, or by random mutagenesis caused by increasing the error rate of PCR of the original polynucleotide with 0.1 mM MnC 1 2 and unbalanced nucleotide concentrations.
  • Nucleic acids used to transfect cells with sequences coding for expression of the polypeptide of interest generally will be in the form of an expression vector that includes expression control sequences operatively linked to a nucleotide sequence coding for expression of the polypeptide.
  • nucleotide sequence “coding for expression of” a polypeptide refers to a sequence that, upon transcription and translation of mRNA, produces the polypeptide. As any person skilled in the art recognizes, this includes all degenerate nucleic acid sequences encoding the same amino acid sequence. This can include sequences containing, e.g., introns.
  • expression control sequences refers to nucleic acid sequences that regulate the expression of a nucleic acid sequence to which it is operatively linked. Expression control sequences are “operatively linked” to a nucleic acid sequence when the expression control sequences control and regulate the transcription and, as appropriate, translation of the nucleic acid sequence.
  • expression control sequences can include appropriate promoters, enhancers, transcription terminators, a start codon (i.e., ATG) in front of a protein-encoding gene, splicing signals for introns, maintenance of the correct reading frame of that gene to permit proper translation of the mRNA, and stop codons.
  • the recombinant nucleic acid can be incorporated into an expression vector comprising expression control sequences operatively linked to the recombinant nucleic acid.
  • the expression vector can be adapted for function in prokaryotes or eukaryotes by inclusion of appropriate promoters, replication sequences, markers, etc.
  • the expression vector can be transfected into a host cell for expression of the recombinant nucleic acid.
  • Host cells can be selected for high levels of expression in order to purify the protein.
  • the host cell can be a prokaryotic or eukaryotic cell selected to study the activity of an enzyme produced by the cell.
  • the cell can be, e.g., a cultured cell or a cell in vivo.
  • Transfected host cells useful in this invention include human kidney 293 cell lines such as K695sw, and K695 7171 , glioma cell lines such as HS695, and neuroblastoma cell lines such as SKN695, described in the Experimental section.
  • a ⁇ peptides can be detected by any method known in the art.
  • the method involves an immunoassay employing binding substances specific for the peptides.
  • one can detect A ⁇ peptides by determining their size, e.g., by HPLC or by mass spectrometry.
  • One step of the screening methods of this invention involves measuring the amount of at least one A ⁇ (x- ⁇ 41) peptide, specifically, in a sample.
  • Measuring A ⁇ (x- ⁇ 41) peptides specifically means measuring A ⁇ (x- ⁇ 41) peptides so as to distinguish that molecule from shorter species of A ⁇ , i.e., those species whose carboxy-terminus extends no further than amino acid # 40 of A ⁇ .
  • Specific measurement of A ⁇ (x- ⁇ 41) preferably is performed by the use of binding substances that specifically recognize A ⁇ (x- ⁇ 41) peptides, e.g., binding substances that recognize amino acids of A ⁇ beyond amino acid # 40.
  • Another method of this invention involves screening compounds to determine their ability to alter the production of both A ⁇ (x- ⁇ 41) peptides and total A ⁇ or A ⁇ (x- ⁇ 40) peptides. Such methods can involve the use of binding substances that can distinguish A ⁇ (x- ⁇ 40) peptides from longer species of A ⁇ , such as A ⁇ (x- ⁇ 41) peptides.
  • immunological detection techniques i.e., immunoassays employing binding substances
  • Particularly suitable detection techniques include ELISA, Western blotting, radioimmunoassay, and the like.
  • immunological methods employing a single antibody are also contemplated, for example, radioimmunoassay using an antibody specific for ⁇ 41 forms of A ⁇ , or single antibody ELISA methods. It will be clear that the particular forms of A ⁇ detected by such methods depend upon the particular binding substances employed. For example, binding substances directed to the junction region may detect A ⁇ (x- ⁇ 41) peptides whose amino termini do not extend to amino acid no. 1 of A ⁇ .
  • binding substances directed to the carboxy-terminal end of A ⁇ (x- ⁇ 41) may detect peptides ending at amino acids 41, 42 or 43. Therefore, determining the specificity of the binding substances will assist in determining exactly which A ⁇ (x- ⁇ 41) peptides the assay is detecting.
  • the method to detect A ⁇ (x- ⁇ 41) peptides is an immunoassay involving two antibodies.
  • One antibody is specific for an epitope containing amino acids beyond number 40 in A ⁇ , and another antibody is capable of distinguishing A ⁇ and A ⁇ fragments from other APP fragments which might be found in the sample.
  • antibodies which are monospecific for the junction region of A ⁇ are capable of distinguishing A ⁇ from other APP fragments.
  • the junction region of A ⁇ is centered at amino acid residues 16 and 17, typically spanning amino acid residues ⁇ 13 to ⁇ 28.
  • Such “junction-recognizing” antibodies may be prepared using synthetic peptides having that sequence as an immunogen.
  • a preferred immunoassay technique is a two-site or “sandwich” assay.
  • This assay involves a capture binding substance, usually bound to a solid phase, and a labelled detection binding substance.
  • a ⁇ (x- ⁇ 41) peptides are captured from the sample using a first binding substance specific for A ⁇ (x- ⁇ 41) peptides (usually bound to a solid phase).
  • the capture of A ⁇ (x- ⁇ 41) peptides is detected using a labeled second binding substance specific for A ⁇ .
  • Labeled binding substances include, for example, those directed to the junction region (amino acids ⁇ 13 to ⁇ 28) or binding substances specific for amino-terminal amino acids (1-5 or 1-12).
  • a sandwich assay using an antibody against the junction region can be used to specifically measure A ⁇ and A ⁇ fragments whose amino-terminus begins before amino acid 13 of A ⁇ . Such assays do not recognize p3 or p3 fragments, since those peptides begin at amino acid # 17 of A ⁇ .
  • Antibodies specific for A ⁇ (x- ⁇ 41), i.e., which do not cross react with A ⁇ ( ⁇ 40), are particularly useful in the methods of this invention. These antibodies can be made by immunizing animals with synthetic peptides that include amino acids beyond number 40 of A ⁇ . For example, the synthetic peptide can include amino acids 33-42. A specific example of the production of such an antibody is provided in the Experimental section.
  • the particular peptides measured from among the group of all A ⁇ (x- ⁇ 41) depends on the particular measuring method used.
  • the binding substance can be directed to one or more from among the group of peptides.
  • an antibody raised against amino acids 33-42 of A ⁇ that does not cross react with A ⁇ (1-40) will bind to A ⁇ (x-42). It also may bind to A ⁇ (x-41) and A ⁇ (x-43).
  • the method involves determining the amount of A ⁇ (x- ⁇ 41) having at least amino acids 13-41 of A ⁇ .
  • a ⁇ can be measured using a sandwich assay employing antibodies that recognize the junction region (amino acids 13-26) and antibodies produced by immunization with a hapten having A ⁇ amino acids 33-42.
  • Total A ⁇ can be measured using a capture antibody to the junction region (e.g., the 266 antibody, described herein) and a reporter antibody that should detect virtually all the A ⁇ peptides and A ⁇ fragments, e.g., an antibody raised against amino acids 1-12 of A ⁇ .
  • Another method of measuring the amount of A ⁇ (x- ⁇ 41) in a sample involves pulse-chase procedures.
  • the culture is pulsed with a radioactive label for protein, e.g., a radioactive amino acid such as 35 S methionine.
  • a radioactive label for protein e.g., a radioactive amino acid such as 35 S methionine.
  • the culture is chased without the label.
  • the compound is administered to the cells.
  • the cells are contacted with a binding substance specific for A ⁇ (x- ⁇ 41) and the amount of radioactive label attached to the binding substance is determined.
  • Antibodies specific for A ⁇ can be prepared, e.g., by immunizing an animal with a peptide whose amino acid sequence corresponds with amino acids ⁇ 13 to ⁇ 28 of A ⁇ .
  • Antibodies specific for A ⁇ (x- ⁇ 41) can be prepared, e.g., by immunizing an animal with a peptide whose amino acid sequence corresponds with amino acids 33-42 of A ⁇ .
  • Antibodies specific for A ⁇ (x- ⁇ 40) can be prepared, e.g., by immunizing an animal with a peptide whose amino acid sequence corresponds with amino acids 33-40 or 28-40 of A ⁇ .
  • Antibodies against the junction region are useful for detecting total A ⁇ .
  • Synthetic polypeptide haptens may be produced by the well-known Merrifield solid-phase synthesis technique in which amino acids are sequentially added to a growing chain (Merrifield (1963) J. Am. Chem. Soc . 85:2149-2156). Suitable peptide haptens will usually comprise at least five contiguous residues within A ⁇ and may include more than six residues. The amino acid sequences may be based on the sequence of A ⁇ set forth above.
  • polypeptide hapten may be conjugated to a suitable immunogenic carrier, such as serum albumin, keyhole limpet hemocyanin, or other suitable protein carriers, as generally described in Hudson and Hay, Practical Immunology , Blackwell Scientific Publications, Oxford, Chapter 1.3, 1980.
  • a suitable immunogenic carrier such as serum albumin, keyhole limpet hemocyanin, or other suitable protein carriers, as generally described in Hudson and Hay, Practical Immunology , Blackwell Scientific Publications, Oxford, Chapter 1.3, 1980.
  • An exemplary immunogenic carrier utilized in the examples provided below is ⁇ -CD3 ⁇ antibody (Boehringer-Mannheim, Clone No. 145-2C11).
  • Antibodies specific for the desired epitope may be produced by in vitro or in vivo techniques.
  • In vitro techniques involve exposure of lymphocytes to the immunogens, while in vivo techniques require the injection of the immunogens into a suitable vertebrate host.
  • Suitable vertebrate hosts are non-human, including mice, rats, rabbits, sheep, goats, and the like.
  • Immunogens are injected into the animal according to a predetermined schedule, and the animals are periodically bled, with successive bleeds having improved titer and specificity.
  • the injections may be made intramuscularly, intraperitoneally, subcutaneously, or the like, and an adjuvant, such as incomplete Freund's adjuvant, may be employed.
  • monoclonal antibodies can be obtained by preparing immortalized cell lines capable of producing antibodies having desired specificity.
  • immortalized cell lines may be produced in a variety of ways. Conveniently, a small vertebrate, such as a mouse, is hyperimmunized with the desired immunogen by the method just described. The vertebrate is then killed, usually several days after the final immunization, the spleen cells removed, and the spleen cells immortalized. The manner of immortalization is not critical. Presently, the most common technique is fusion with a myeloma cell fusion partner, as first described by Kohler and Milstein (1975) Nature 256:495-497.
  • the detection techniques of the present invention will also be able to use antibody fragments, such as F(ab), Fv, V L , V H , and other fragments.
  • antibody fragments such as F(ab), Fv, V L , V H , and other fragments.
  • polyclonal antibodies it may be necessary to adsorb the anti-sera against the target epitopes in order to produce a monospecific antibody population.
  • recombinantly produced antibodies immunoglobulins
  • kits for performing assays of the invention include means for detecting specifically A ⁇ (x- ⁇ 41) and means for detecting specifically A ⁇ (x- ⁇ 40).
  • the means can include any means known or described above, e.g., binding substances.
  • the kit includes a binding substance specific for A ⁇ (x- ⁇ 41) (i.e., that does not cross react with A ⁇ ( ⁇ 40)), and a binding substance specific for A ⁇ ( ⁇ 40) (i.e., that does not cross react with A ⁇ (x- ⁇ 41)).
  • a binding substance specific for A ⁇ (x- ⁇ 41) i.e., that does not cross react with A ⁇ ( ⁇ 40)
  • a binding substance specific for A ⁇ ( ⁇ 40) i.e., that does not cross react with A ⁇ (x- ⁇ 41)
  • the kit is useful for immunoassays including two antibodies for each antigen.
  • the kit can further comprise a binding substance specific for the junction region of A ⁇ .
  • Such antibodies are useful for the capture or detection of both A ⁇ (x- ⁇ 41) and A ⁇ ( ⁇ 40).
  • the binding substance specific for the junction region is bound to a solid phase, and the binding substances specific for A ⁇ (x- ⁇ 41) and A ⁇ ( ⁇ 40) are detectably labeled.
  • the detectable labels can be any known and used in the art including, e.g., a biotinylation label, a radioactive label, a light scattering label, an enzymatic label, a fluorescent label and the like.
  • the kit can further comprise a substrate for the enzyme.
  • test compounds can be any molecule, compound, or other substance which can be added to the cell culture or administered to the test animal without substantially interfering with cell or animal viability.
  • Suitable test compounds may be small molecules (i.e., molecules whose molecular mass is no more than 1000 Daltons), biological polymers, such as polypeptides, polysaccharides, polynucleotides, and the like.
  • the test compounds will typically be administered to the culture medium at a concentration in the range from about 1 nM to 1 mM, usually from about 10 ⁇ M to 1 mM.
  • the test compounds will typically be administered at a dosage of from 1 ng/kg to 100 mg/kg, usually from 10 ⁇ g/kg to 1 mg/kg.
  • Test compounds which are able to inhibit secretion or production of A ⁇ (x- ⁇ 41) are considered as candidates for further determinations of the ability to block ⁇ -amyloid production in animals and humans. Such compounds can be tested in in vivo studies, as described below. Inhibition of secretion or production indicates that cleavage of A ⁇ between amino acids 42/43 has likely been at least partly blocked, reducing the amount of A ⁇ (x- ⁇ 41) available for forming ⁇ -amyloid plaques.
  • a preferred non-human transgenic animal is one whose cells harbor a PDAPP construct.
  • a PDAPP construct is a nucleic acid construct that comprises a mammalian promoter operatively linked to a cDNA-genomic DNA hybrid coding for the expression of APP.
  • the cDNA-genomic DNA hybrid contains a cDNA sequence encoding APP770 or a cDNA sequence encoding APP770 with a naturally occurring mutation (e.g., a Hardy mutation or the Swedish mutation) substituted with genomic DNA sequences.
  • the genomic DNA sequences consist of exon 6 and an amount of the adjacent downstream intron sufficient for splicing, the KI and OX-2 coding region and an amount of each of their upstream and downstream introns sufficient for splicing, and exon 9 and an amount of the adjacent upstream intron sufficient for splicing, substituted into the corresponding region of the cDNA sequence encoding APP770, or the cDNA encoding APP770 with a naturally occurring mutation.
  • the construct is transcribed and differentially spliced in mammalian cells to form mRNA molecules that encode and that are translated into APP695, APP751 and APP770.
  • the construct contains a PDGF- ⁇ promoter operatively linked with a hybrid sequence encoding an APP gene harboring a Hardy mutation (V717F), and the SV40 polyadenylation signal.
  • V717F Hardy mutation
  • V717F Hardy mutation
  • SV40 polyadenylation signal One version of the PDAPP construct is presented in Example IX.
  • Another useful non-human animal model harbors a copy of an expressible transgene sequence which encodes the Swedish mutation of APP (asparagine 595 -leucine 596 ).
  • the sequence generally is expressed in cells which normally express the naturally-occurring endogenous APP gene (if present).
  • transgenes typically comprise a Swedish mutation APP expression cassette, in which a linked promoter and, preferably, an enhancer drive expression of structural sequences encoding a heterologous APP polypeptide comprising the Swedish mutation.
  • a ⁇ levels can be measured in any body fluid or tissue sample, for example, brain homogenate.
  • the transgenic animals that are usually produced by introducing the transgene or targeting construct into a fertilized egg or embryonic stem (ES) cell, typically by microinjection, electroporation, lipofection, or biolistics.
  • the transgenic animals express the Swedish mutation APP gene of the transgene (or homologously recombined targeting construct), typically in brain tissue.
  • ES embryonic stem
  • one or both endogenous APP alleles is inactivated and incapable of expressing the wild-type APP.
  • test compounds can be administered to additional test animals, in which deviation from the average control value would indicate that the test compound had an effect on the ⁇ -secretase activity in the animal.
  • Test substances which are considered positive, i.e., likely to be beneficial in the treatment of Alzheimer's disease or other ⁇ -amyloid-related conditions will be those which are able to reduce the level of A ⁇ (x- ⁇ 41) production, preferably by at least 20%, more preferably by at least 50%, and most preferably by at least 80%.
  • junction peptide Monoclonal antibodies to the junction region of A ⁇ were prepared using a synthetic peptide spanning amino acid residues 13-30, except that AI, amino acids 30 and 31, were substituted with GC (the “junction peptide”).
  • the junction peptide was conjugated to an immunogen ( ⁇ -CD3 ⁇ antibody; Clone No. 145-2C11, Boehringer-Mannheim) using m-maleimidobenzoyl-N-hydroxysuccinimide ester (MHS) according to the manufacturer's (Pierce) instructions.
  • A/J mice were immunized initially intraperitoneally (IP) with the A ⁇ conjugate mixed with complete Freund's adjuvant. Fourteen days later, the mice were boosted IP with the A ⁇ conjugate mixed with phosphate buffered saline (PBS) at 14 day intervals. After six total boosts, the mice were finally boosted intravenously with A ⁇ conjugate mixed with incomplete Freund's adjuvant and fused 3 days later. Fusion of spleen cells with P3.653 myeloma cells was performed as described in Oi and Herzenberg, Selective Methods in Cellular Immunology , Mishell and Shigii, Eds., W. H. Freeman and Company, San Francisco, Chapter 17 (1980). Serum titers and initial screens were performed by the RIA method described below. Several clones were expanded to a 24-well plate and subjected to further analysis as described below. Clones of interest were produced in mouse ascites.
  • the RIA method used to screen serum bleeds and fusion hybridoma supernatants was based upon a method developed by Wang et al. (1977) J. Immunol. Methods 18:157-164. Briefly, the supernatant (or serum) was incubated overnight at room temperature on a rotator with 125 I-labeled A ⁇ 1-28 and Sepharose® 4B beads to which sheep anti-mouse IgG had been coupled via cyanogen bromide. The beads from each well were harvested onto glass fiber filter discs with a cell harvester and washed several times with PBS. The filter discs were then transferred to gamma tubes and the bound radioactivity was counted in a gamma counter.
  • the clones were also characterized based on reactivity in Western blots. Based on titer point, sensitivity (as determined by the 50% inhibition point), and reactivity on Western blot, several clones were produced in ascites. Antibodies from hybridoma designated 266 (the “266 antibody”) was selected for use as a capture antibody in the assays described below.
  • Antibody 2G3, specific for A ⁇ (x-40), was produced by injecting female A/J mice intraperitoneally with 100 ⁇ g immunogen per injection.
  • the immunogen consisted of the peptide NH 2 -Cys-NH—CH 2 —(CH 2 ) 5 —CO-GLMVGGVV-COOH (SEQ ID NO: 5), coupled to sheep anti-mouse IgG using maleimidohexanoyl-N-hydroxysuccinimide.
  • the immunogen was emulsified with Freund's complete adjuvant for the first immunization, and all subsequent immunizations were with 100 ⁇ g of immunogen emulsified with Freund's incomplete adjuvant at approximately two week intervals.
  • a mouse was boosted with PBS solutions containing 50 ⁇ g immunogen intravenously and 50 ⁇ g intraperitoneally in PBS.
  • the mouse was sacrificed, the spleen was removed, splenocytes were isolated and fused with the SP2/0 mouse myeloma using a modification of the method of Koehler and Milstein.
  • Antibody 21F12 specific for A ⁇ (x ⁇ 41) was produced by immunizing A/J mice intraperitoneally with 100 ⁇ g of immunogen per injection.
  • the immunogen consists of the synthetic peptide NH 2 -Cys-NH—CH 2 (CH 2 ) 5 —CO-GLMVGGVVIA-COOH (SEQ ID NO: 4) coupled to sheep anti-mouse IgG using maleimidohexanoyl-N-hydroxysuccinimide (MHS).
  • MHS maleimidohexanoyl-N-hydroxysuccinimide
  • a mouse was injected with 50 ⁇ g of immunogen each intravenously and intraperitoneally of immunogen in PBS.
  • the spleen was removed, splenocytes were isolated and fused with SP2/0 following a modification of the method of Koehler and Milstein.
  • antibody 2G3 and 21F12 The specificities of antibody 2G3 and 21F12 are demonstrated in FIGS. 1 and 2 .
  • antibody 2G3 or 21F12 was coated into the wells of an ELISA plate by diluting the purified antibody to a concentration of 10 ⁇ g/ml in Well-coating Buffer (0.01 M PO 4 pH 8.5), and pipetting 100 ⁇ l of the antibody solution into each well. The solution was left overnight at room temperature and then was removed by aspiration. The non-specific sites of the well were blocked by the addition of 200 ⁇ l 0.25% Human Serum albumin in PBS and incubated for at least one hour at room temperature. The blocking solution was removed and the wells were washed one time with wash buffer (Tris buffered Saline, 0.05% Tween 20).
  • wash buffer Tris buffered Saline, 0.05% Tween 20
  • Standards containing between 80-20,000 pg/ml of either A ⁇ (1-40) or A ⁇ (1-42) were then prepared by dilution in Specimen Diluent (1 mM PO 4 , 0.15 M NaCl, pH 7.4, 0.6% Bovine Serum albumin, globulin-free, 0.05% Triton X-405 and 0.05% Thimerosal), and 100 ⁇ l of each of these standards were added to the appropriate wells. The standards were incubated for one hour at room temperature, then aspirated and the wells washed four times with wash buffer.
  • reporter antibody 100 ⁇ l of a second antibody (the reporter antibody) was added at a concentration of 0.5 ⁇ g/ml in specimen diluent.
  • This reporter antibody is biotinylated 3D6 (which recognizes A ⁇ (1-5)) prepared by the reaction of antibody with NHS-biotin (Pierce). This was allowed to incubate one hour at room temperature, and then washed four times with wash buffer.
  • antibody 2G3 reacts strongly with A ⁇ (1-40), but has essentially no cross-reactivity with A ⁇ (1-42).
  • FIG. 2 it is shown that antibody 21F12 similarly has very high specificity, in this case for A ⁇ (1-42) over A ⁇ (1-40). At a concentration of 20,000 pg/ml less than 0.4% of cross reactivity is observed.
  • a monoclonal antibody against A ⁇ (x ⁇ 41) or A ⁇ (x ⁇ 40) is diluted to a concentration of 10 ⁇ g/ml in a buffer containing 0.23 g/L NaH 2 PO 4 .H 2 O, 26.2 g/L Na 2 HPO 4 .7H 2 O, 1 g/L NaN 3 , pH 8.5.
  • One hundred ⁇ l/well of this solution is then dispensed in a 96 well white Dynatech Microlite 2, 96 well flat-bottomed plate. The plates are sealed and incubated overnight at room temperature.
  • the calibrators are prepared from a stock solution of A ⁇ 1-42 , 1 ⁇ g/ml, in DMSO.
  • specimen diluent ((NaH 2 PO 4 .H 2 O) 0.2 g/L, Na 2 HPO 4 .7H 2 O 2.16 g/L, NaN 3 0.5 g/L, bovine serum albumin (BSA) (globulin free) 6 g/L, triton x-405 0.5 ml/L NaCl 8.5 g/L, pH 7.4.)
  • the highest calibrator 1000 pg/ml (10 ⁇ l A ⁇ 1-42 stock (1 ⁇ g/ml DMSO) in 10 ml casein specimen diluent) is prepared. Sequential dilutions are made in specimen diluent to obtain 500, 250, 125, 62.5 and 31.25 pg/ml concentrations of A ⁇ 1-42 .
  • washing buffer NaCl 80 g/L, KCl 3.85 g/L, Tris-HCl 31.75 g/L, tween-20 0.5 ml/L, pH 7.5.
  • Antibody is diluted in specimen diluent to 1 ⁇ g/ml and 100 ⁇ l is added per well. The plate is covered and incubated for 1 hour at room temperature. The plate is washed three times with washing buffer. The alkaline phosphatase affinity purified F(ab′)2 fragment donkey anti-rabbit IgG (H+L) (Jackson) is diluted 1:1000 in specimen diluent. One hundred ⁇ l/well is added. The plate is covered and incubated for 1 hour at room temperature. The plate is washed three times with washing buffer, then 100 ⁇ l/well of chemiluminescent substrate is added.
  • the chemiluminescent substrate is prepared by diluting the chemiluminescent reagent, AMPPD (Tropix), and an enhancer, emerald green (Tropix), 1:1000 and 1:100 respectively in 1M diethanolemine buffer, pH 10, containing 1 mM MgCl 2 and 0.2% NaN 3 .
  • the plates are sealed and incubated for 10 to 15 minutes at room temperature. Solution is not aspirated. This time may have to be optimized for different antibody lots.
  • Chemiluminescence is read and expressed as relative chemiluminescence units (CLU) after 15 minutes using a Dynatech ML 1000.
  • MDL 28170 Inhibits the Production of A ⁇ (40) and p3(40) but not A ⁇ (42) and p3(42)
  • MDL 28170 had previously been shown to inhibit the secretion of A ⁇ and p3 and therefore had been suggested to inhibit ⁇ -secretase. This inhibition was observed by immunoprecipitating media from treated cells with a polyclonal antibody raised to synthetic A ⁇ (1-40) (Higaki et al., (1995) Neuron , 14:651-659). Since the vast majority of secreted A ⁇ and p3 peptides end at amino acid 40, this experiment does not distinguish whether only the major ⁇ -secretase cleavage at position 40 is inhibited.
  • a ⁇ (42) and p3(42) are not decreased by MDL 28170 depends critically on the quality of the 21F12 antibody.
  • two other previously published A ⁇ (42)-specific antibodies were used in the pulse chase paradigm with MDL 28170 at 200 ⁇ M.
  • the monoclonal antibody BC05 has been extensively used in ELISA assays to detect A ⁇ (42) (Asami-Odaka et al., (1995) Biochemistry 34:10272-10278; Gravina et al., (1995) J. Biol. Chem . 270:7013-7016; Suzuki et al., (1994) Science 264:1336-1340).
  • the polyclonal antibody C42 has also been shown to be specific for A ⁇ (42) (Saido et al., (1994) Spatial resolution of the primary ⁇ -amyloidogenic process induced in postischemic hippocampus. J. Biol. Chem . 269:15253-15257). Likewise, this antibody did not show a decrease in A ⁇ (42) and p3(42) upon treatment whereas the subsequent precipitation with R1282 showed the usual decrease in total A ⁇ and p3 ( FIG. 5C ).
  • a ⁇ (40) and p3(40) were also found when the monoclonal antibody, 2G3 (described above) specific for the free carboxyl-terminus of A ⁇ (40) and p3(40) was used to precipitate first, followed by 21F12 ( FIG. 5D ).
  • the differential inhibition of A ⁇ production by MDL 28170 was also detected when the precipitations were carried out not sequentially (as described above) but in parallel after the standard pulse-chase. That is, aliquots of media from treated cells and from untreated cells were precipitated with 21F12 for the A ⁇ 42 forms and other aliquots were precipitated with antibody 1282 for total A ⁇ and total p3. This parallel precipitation produced the same result as the sequential precipitations described above ( FIG. 5E ).
  • kidney cell line K695 717I expresses APP 695 carrying the 717I mutation. This line was chosen because it produces increased levels of A ⁇ (42) due to the mutation (Suzuki et al., (1994) Science 264:1336-1340). At 200 ⁇ M MDL 28170, no decrease of A ⁇ (42) and p3(42) was observed, whereas A ⁇ (40) and p3(40) were substantially reduced. ( FIG. 6A ).
  • the Chinese Hamster ovary cell line CHO695 transfected with wild-type ⁇ APP 695 cDNA was treated with 200 ⁇ M MDL 28170 and only a slight decrease of A ⁇ (42) and p3(42) was observed, whereas A ⁇ (40) and p3(40) were substantially reduced.
  • FIG. 6B The human neuroblastoma cell-line SKN695 expressing wild type ⁇ APP 695 was treated with 200 ⁇ M MDL 28170 ( FIG. 6C ). While A ⁇ (42) and p3(42) were slightly increased, total A ⁇ and total p3 were strongly decreased.
  • differential inhibition of A ⁇ (42) vs A ⁇ (40) and p3(42) vs p3(40) production is not only observed in K695sw but also in a cell line with an Alzheimer's disease linked ⁇ APP717 missense mutation, in a hamster cell line and in a human neural cell line expressing wild type ⁇ APP.
  • K695sw are human embryonic kidney 293 cells stably transfected with a construct carrying the AD-linked double (“Swedish”) mutation K595N/M596L (Citron et al. (1992) Nature , 360:672-674); K695 717I are 293 cells stably transfected with APP 695 carrying the mutation V717I (valine to isoleucine at position 717 in the APP770 numbering system).
  • CHO695 are Chinese hamster ovary cells (CHO) stably transfected with pCMV695 (Oltersdorf (1990) J. Biol. Chem . 265:4432-4437)
  • SKN695 are SK-N-SH human neuroblastoma cells stably transfected with pCMV695.
  • MDL 28170 To analyze the effect of MDL 28170 on the processing of APP, cells were grown to confluence in two 10 cm dishes, pulse-labeled with 600 ⁇ Ci of [ 35 S]-methionine in 4 ml of serum-free medium for 2 hours and then chased for 2 hours with 4 ml medium containing 10% fetal bovine serum and the indicated final concentration of MDL 28170 (initially dissolved at 200 mM. in DMSO). Control dishes were treated with DMSO alone.
  • This antibody precipitates total A ⁇ and p3 (and small, variable amounts of APP s ) from the media of cultured cells (Haass, et al. (1992) Nature 359:322-325).
  • the monoclonal antibody 2G3 was raised to peptide C(Aminoheptanoic acid)GLMVGGVV [(SEQ ID NO: 5) and specifically precipitates A ⁇ (40) and p3(40). Twenty ⁇ g of this antibody were used to immunoprecipitate the chase media of 2 dishes.
  • the monoclonal antibody 21F12 was raised to peptide C(Aminoheptanoic acid)GLMVGGVVIA (SEQ ID NO: 4) and specifically precipitates A ⁇ (42) and p3(42).
  • the monoclonal antibody BC05 specifically detects A ⁇ (42) and p3(42) (Suzuki et al., (1994) Science 264:1336-1340).
  • the polyclonal antibody C7 against the last 20 residues of the APP cytoplasmic tail (Podlisny, (1991) Am. J. Pathol. 138:1423-1435) precipitates N′- and N ⁇ plus O ⁇ -glycosylated full-length APP as well as its C-terminal proteolytic fragments.
  • the antibody sw192 Karl et al. (1995) J. Biol. Chem. 270:2419-2422; Haass et al. (1995) Nature Med. 1:1291-1296
  • Human neurons were cultured as previously described (Seubert et al. Nature (1992) 359:325-327) except that the cells were seeded into 6-well plates in neuronal medium without fetal bovine serum but supplemented with B27 (Gibco). Cells were cultured for 2-3 weeks in serum free medium prior to use.
  • PDAPP mouse brain cells from 16 day old fetal cerebral cortex were cultured following the protocol for human neurons except the cells were seeded into 24-well plate clusters in neuronal medium with 50 fetal bovine serum (Sigma) and 5% Chang's supplement (Irvine Scientific). Cells were cultured for 5-7 days prior to being used in experiments.
  • Fresh medium is added to the culture wells and then collected after ⁇ 24 (8-30) hrs. This is the “control” sample from each well that the treated sample will be compared to.
  • Fresh medium, containing the substance to be tested is then added and again harvested after a further ⁇ 24 hr (8-30) incubation.
  • a cytotoxicity assay is performed on the cells. To perform the cytotoxicity assay, cells are incubated in media containing thiazolyl blue (MTT, Sigma) at 1 mg/ml for 15 minutes. The media are then discarded, and the precipitates are analyzed by solubilization in a buffer containing 50% DMF and 20% SDS. The solubilized dye was quantitated on a Molecular Devices Vmax.
  • Control and treated samples of culture media are assayed for total A ⁇ using a sandwich ELISA consisting of two monoclonal antibodies.
  • the first antibody 266, specific to amino acids 13-28 of A ⁇ , is used as a capture antibody (Seubert et al., Nature , supra).
  • the second antibody, 3D6 which is specific to amino acids 1-5 of A ⁇ , was biotinylated and served as a reporter antibody.
  • the 3D6 biotinylation procedure employed the manufacturer's protocol for NHS-biotin (Pierce) labeling of immunoglobulins, except 100 mM sodium bicarbonate, pH 8.5, buffer was used.
  • the 3D6 antibody does not recognize secreted APP or full-length APP but does recognize A ⁇ species that begin at position 1.
  • the samples were also assayed for A ⁇ (42) with an A ⁇ (42) specific sandwich ELISA that employed the monoclonal antibody 21F12, which was generated against amino acids 33-42 of A ⁇ , as the capture antibody.
  • This antibody is specific for longer forms of A ⁇ since it does not cross-react with A ⁇ (1-40) in ELISA or competitive radioimmunoassay (RIA).
  • Biotinylated 3D6 is also the reporter antibody in this assay.
  • the 266 and 21F12 mAbs were coated at 10 ⁇ g/ml into 96-well immunoassay plates (Costar) overnight at room temperature.
  • the plates were aspirated and blocked with 0.25% human serum albumin PBS buffer for at least 1 hour at room temperature, then stored desiccated at 4° C. until use.
  • the samples and standards were added to the plates and incubated at room temperature for 1.5 hours.
  • the biotinylated 3D6 was diluted to 0.5 ⁇ g/ml, and incubated in the wells for 1 hour at room temperature.
  • the plates were washed 3 times with wash buffer (Tris buffered saline, 0.05% Tween 20) between each step of the assay.
  • Streptavidin-alkaline phosphatase (Boehringer Mannheim), diluted 1:1000, was added to the wells for the total A ⁇ assay, and avidin-HRP (Vector) diluted 1:4000 was added to the wells for the A ⁇ (42) assay. These conjugates were incubated for 1 hour at room temperature.
  • the fluorometric substrate 4-methyl-umbelliferyl phosphate was added to the wells for 30 minutes, then read in a Millipore Cytofluor 2350 fluorometer.
  • the calorimetric substrate, Slow TMB-ELISA (Pierce) was added for the A ⁇ (42) assay and allowed to react for 15 minutes, after which the enzymatic reaction was stopped with 2N H 2 SO 4 .
  • the plates were read on a Molecular Devices Vmax.
  • a cDNA/genomic APP construct containing introns 6, 7 and 8 is prepared by combining APP cDNA encoding exons 1-6 and 9-18 with genomic APP sequences encoding introns 6, 7 and 8, and exons 7 and 8 (see FIGS. 8A-8B ).
  • two deletions in intronic sequences are made.
  • a deletion is made in intron 6 from position 143 of intron 6 to the BamHI site located upstream of the beginning of exon 7 (1658 bp before the beginning of exon 7).
  • Another deletion is made in intron 8 from the first BamHI site in intron 8 to a site at 263 bp before the beginning of exon 9.
  • intron ⁇ 6 and ⁇ 8 these truncated forms of APP introns 6 and 8 are referred to herein as intron ⁇ 6 and ⁇ 8.
  • BamHI sites are engineered at the sites of these deletions, so that they are marked by the presence of BamHI sites.
  • exons 7 and 8 and intron 7 are intact genomic sequences, except that the unique XhoI site in intron 7 is destroyed.
  • DNA fragments containing the truncated introns are generated as follows: a BamHI site is engineered 143 bp into intron 6 nucleotide by PCR-mutagenesis (“Mutagenesis by PCR” in PCR Technology: Current Innovations (Griffith and Griffith, eds., CRC Press, 1994) pages 69-83) and another BamHI site is engineered by PCR mutagenesis 263 bp prior to the beginning of exon 9. These sites are engineered into separate APP genomic DNA clones containing the junctions of exon 6 and intron 6, and intron 8 and exon 9, respectively, resulting in modified APP genomic DNA clones.
  • the entire cassette is assembled in the APP cDNA clone as follows ( FIG. 9 ).
  • the 889 bp BamHI to XcmI fragment of APP cDNA containing exons 1 through 5 and part of exon 6 (including nucleotides 1 to 843 of FIG. 10 (SEQ ID NO: 2)) is cloned into a vector containing BamHI and XhoI sites downstream from the insertion site to make APP770x-oligo-x.
  • APP770x-oligo-x is then cut with XcmI and BamHI.
  • two fragments are obtained from the modified APP genomic DNA clone containing the junction of exon 6 and intron 6 described above by cutting with XcmI and BamHI.
  • the resulting 34 bp fragment from the XcmI in exon 6 to the XcmI in intron 6, and 131 bp fragment from the XcmI in intron 6 to the artificially created BamHI site at position 143 bp of intron 6 are ligated into APP770x-oligo-x in a three-way ligation step to make APP 770x-E6oligo-x.
  • the orientation of the fragments are confirmed by sequencing.
  • APP770x-E6oligo-x is then cut with BamHI and XhoI.
  • APP770xE6E9x is then cut with BamHI and the 6.8 kb BamHI fragment of APP genomic DNA encoding the KPI and OX-2 domains (exons 7 and 8) is inserted at this site.
  • This fragment starts at the BamHI site 1658 bp upstream of the start of exon 7 and extends to the first BamHI site in intron 8.
  • This BamHI fragment is obtained from a lambda phage genomic clone encoding this portion of the APP gene, that was obtained from a Human Placental genomic library in the Lambda FIXII vector obtained from Stratagene.
  • This BamHI fragment originally contained an XhoI site which was destroyed by cutting fill in and relegation.
  • This clone containing exons 1-8 and part of 9, and introns 6, 7 and 8, is termed the “APP splicing cassette.”
  • the APP splicing cassette is cut out with NruI and XhoI and used to replace the NruI to XhoI cDNA fragment of APP cDNA bearing a Hardy mutation.
  • This mutant form of APP cDNA is produced by converting the G at nucleotide position 2145 to T by site directed mutagenesis. This changes the encoded amino acid from Val to Phe.
  • the resulting construct is a combination cDNA/genomic APP “minigene.”
  • the 1.60 kb EcoRI fragment containing exon 8 is actually upstream of the 1.48 kb EcoRI fragment and the 1.48 kb EcoRI fragment Yoshikai et al. mapped into intron 7 is actually in intron 8.
  • This APP minigene is operatively linked to the PDGF- ⁇ promoter to provide expression of the APP cDNA/genomic construct in mammalian cells.
  • the PDGF ⁇ -chain 5′ flanking sequence is inserted upstream of the NruI site at the beginning of the APP minigene.
  • This fragment includes 1.3 kb upstream of the transcription initiation site, where the PDGF- ⁇ promoter resides, and approximately 70 bp of 5′ untranslated region, ending at the AurII site (Higgins et al. (1994)).
  • the late SV40 polyadenylation signal carried on a 240 bp BamHI to BclI fragment, is added downstream of the APP minigene.
  • This construct, combining the PDGF- ⁇ promoter, the APP splicing cassette, a Hardy mutation, and the SV40 polyadenylation signal is referred to as PDAPP ( FIG. 12 ).
  • the present invention provides a novel screening method for determining whether a compound alters the production of A ⁇ (x- ⁇ 41) and/or A ⁇ (x- ⁇ 40) or total A ⁇ . While specific examples have been provided, the above description is illustrative and not restrictive. Many variations of the invention will become apparent to those of skill in the art upon review of this specification. The scope of the invention should, therefore, be determined not with reference to the above description, but instead should be determined with reference to the appended claims along with their full scope of equivalents.

Abstract

This invention provides methods of screening compounds for their ability to alter the production of Aβ(x≧41) alone or in combination with Aβ(x≦40). The methods involve administering compounds to cells, specifically measuring the amounts of Aβ(x≦40) and Aβ(x≧41) produced by the cells, and comparing these amounts to that produced by the cells without administration of the compounds.

Description

This application is a divisional of U.S. application Ser. No. 08/665,649 filed Jun. 18, 1996 which is now U.S. Pat. No. 6,610,493.
This invention was made in part with Government support under grant no. 2 P50 AG05134 awarded by the National Institutes of Health. The Government may have rights in this invention.
BACKGROUND OF THE INVENTION
1. Field of the Invention
The present invention relates generally to neurology and, more particularly, to assays, such as immunoassays, for screening for compounds that specifically alter the production of various isoforms of Aβ.
Alzheimer's disease (AD) is a degenerative brain disorder characterized clinically by progressive loss of memory, cognition, reasoning, judgment and emotional stability that gradually leads to profound mental deterioration and ultimately death. AD is a very common cause of progressive mental failure (dementia) in aged humans and is believed to represent the fourth most common medical cause of death in the United States. AD has been observed in all races and ethnic groups worldwide and presents a major present and future public health problem. The disease is currently estimated to affect about two to three million individuals in the United States alone. AD is at present incurable. No treatment that effectively prevents AD or reverses its symptoms or course is currently known.
The brains of individuals with AD exhibit characteristic lesions termed senile plaques and neurofibrillary tangles. Large numbers of these lesions are generally found in patients with AD in several areas of the human brain important for memory and cognitive function. Smaller numbers of these lesions in a more restricted anatomical distribution are sometimes found in the brains of aged humans who do not have clinical AD. Senile plaques and vascular amyloid deposits (amyloid angiopathy) also characterize the brains of individuals beyond a certain age with Trisomy 21 (Down's Syndrome) and Hereditary Cerebral Hemorrhage with Amyloidosis of the Dutch-Type (HCHWA-D). The principal chemical constituent of the senile plaques and vascular amyloid deposits characteristic of AD and the other disorders mentioned above is a protein designated the amyloid-β peptide (Aβ) or sometimes βAP, AβP or β/A4. Aβ was first purified and a partial amino acid sequence reported in Glenner and Wong (1984) Biochem. Biophys. Res. Commun.120:885-890. The isolation procedure and the sequence data for the first 28 amino acids are described in U.S. Pat. No. 4,666,829. Forms of Aβ having amino acids beyond number 40 were first reported by Kang et al. (1987) Nature 325:733-736.
Roher et al. (1993) Proc. Natl. Acad. Sci. USA 90:10836-840 showed that Aβ(1-42) is the major constituent in neuritic plaques, including significant amounts of isomerized and racemized aspartyl residues as their NH2-termini. The authors also reported that Aβ(17-42) (p3(42)) predominates in diffuse (early) plaques, whereas Aβ(1-40) is the major constituent in the meningeal vessel deposits, comprising 60% of the total Aβ in those vessels. Iwatsubo et al. (1994) Neuron 13:45-53 showed that Aβ42(43)-containing senile plaques are the major species of senile plaques in sporadic AD brains. Iwatsubo et al. (1995) Annals of Neurology 37:294-299 and Lemere et al. (1996) Neurobiology of Disease 3:16-32 reported that Aβ42(43) is the major constituent of senile plaques in Down's syndrome brains and is the initially deposited Aβ species in the development of AD-type neuropathological legions in these patients. In addition, Gravina et al., (1995) J. Biol. Chem. 270:7013-7016 reported both biochemical and immunocytochemical evidence that Aβ42(43) peptides were the most abundant constituents of senile plaques in AD brains and exceeded the amounts of Aβ40 peptides in such plaques.
Molecular biological and protein chemical analyses conducted during the last several years have shown that Aβ is a small fragment of a much larger precursor protein, referred to as the β-amyloid precursor protein (APP), that is normally produced by cells in many tissues of various animals, including humans. Knowledge of the structure of the gene encoding APP has demonstrated that Aβ arises as a peptide fragment that is cleaved from the carboxy-terminal end of APP by as-yet-unknown enzymes (proteases). The precise biochemical mechanism by which the Aβ fragment is cleaved from APP and subsequently deposited as amyloid plaques in the cerebral tissue and in the walls of cerebral and meningeal blood vessels is currently unknown. Importantly, Haass et al. (Nature 359:322-325) and Seubert et al. ((1992) Nature 359:325-327) discovered that essentially all cells expressing the APP gene normally secrete an array of Aβ peptides, and these peptides can readily be detected and assayed in cell culture fluid (conditioned media) and human biological fluids such as plasma and cerebrospinal fluid. It has subsequently been shown that these fluids contain both the more abundant Aβ40-ending peptides and the less abundant Aβ42(43)-ending peptides (Dovey et al. (1993) Neuroreport 4:1039-1042 and Vigo-Pelfrey et al. (1993) J. Neurochem. 61:1965-68)
Several lines of evidence indicate that progressive cerebral deposition of Aβ plays a seminal role in the pathogenesis of AD and can precede cognitive symptoms by years or decades (for review, see Schenk (1995) J. Med. Chem. 38:4141-4154, Selkoe (1994) J. Neuropath. and Exp. Neurol. 53:438-447 and Selkoe (1991) Neuron 6:487). One of the most important lines of evidence is the discovery in 1991 that missense DNA mutations in the APP gene at amino acid 717 of the 770-amino acid isoform of APP can be found in affected members but not unaffected members of several families with a genetically determined (familial) form of AD (Goate et al. (1991) Nature 349:704-706; Chartier Harlan et al. (1991) Nature 353:844-846; and Murrell et al. (1991) Science 254:97-99). Suzuki et al. (1994) “An increased percentage of long amyloid β-protein secreted by familial amyloid β-protein precursor (βAPP717) mutants,” Science 264:1336-1340 subsequently showed that, compared to normal individuals, the 717 mutation causes a higher relative production of the Aβ(1-42) peptide. In addition, a double mutation changing lysine670-methionine671 to asparagine670-leucine671 (with reference to the 770 isoform of APP) was reported in a Swedish family with familial AD in 1992 (Mullan et al. (1992) Nature Genet 1:345-347) and is referred to as the Swedish APP variant.
Genetic linkage analyses have demonstrated that the aforementioned mutations are the specific molecular cause of AD in the members of such families that carry these mutant APP genes. In addition, a mutation at amino acid 693 of the 770-amino acid isoform of APP has been identified as the cause of the Aβ deposition disease, Hereditary Cerebral Hemorrhage With Amyloidosis Dutch type (HCHWA-D), and a mutation from alanine to glycine at amino acid 692 appears to cause the phenotype of AD in some family members and the phenotype of HCHWA-D in others. The discovery of these APP mutations in genetically based cases of AD argues that genetic alteration of APP and subsequent deposition of its Aβ fragment can cause AD.
Recently, evidence has accumulated suggesting that Aβ(42) plays the key role in the process of senile plaque formation in AD. First, in vitro data demonstrate that Aβ(42) accelerates the formation of Aβ fibrils (and thus senile plaques) by a nucleation dependent mechanism (Jarrett et al. (1993) Biochemistry 32:4693-4697). Second, while accounting for ≦10% of total Aβ secreted from cells (roughly 90% is Aβ(40) (Dovey et al. (1993) Neuroreport 4:1039-1042; Asami-Odaka et al. (1995) “Long amyloid β-protein secreted from wild-type human neuroblastoma IMR-32 cells.” Biochemistry 34:10272-10278), Aβ(42) is the major plaque component (Kang et al. (1987) Nature 325:733-736; Iwatsubo et al. (1994) Neuron 13:45-53; Iwatsubo et al. (1995) Ann. Neurol. 37:294-299; Gravina et al. (1995) J. Biol. Chem. 270:7013-7016; Lemere et al. (1996) Neurobiology of Disease 3:16-32). Furthermore, all 3 early onset familial AD genes identified to date have been shown to lead to an increase in cellular secretion of Aβ(42). Only the Swedish APP missense mutation increases the secretion of both Aβ(40) and Aβ(42) peptides (Dovey et al. (1993) Neuroreport 4:1039-1042, whereas the APP717 mutations and the presenilin mutations appear not to increase Aβ(40) peptides (Suzuki et al., (1994) Science 264:1336-1340; Scheuner et al. (1995) Neurosci. Abstracts in press). Thus, the longer Aβ(42) peptide appears to be a prime target for therapeutic intervention. However, none of the proteases involved in the major steps of APP processing have been definitively identified, including γ-secretase, the protease which generates the C-terminus of Aβ. It has generally been assumed that the same protease(s) generate both Aβ(40) and A(42) and it has been shown that both forms share a common secretory mechanism which involves acidic intracellular compartments such as the late Golgi or early endosomes (Koo and Squazzo (1994) J. Biol. Chem. 269:17386-17389; Asami-Odaka et al. (1995) Biochemistry 34:10272-10278). Recently, Higaki et al. ((1995) Neuron, 14:651-659) have shown that the Calpain inhibitor, MDL 28170, inhibits the production of both total Aβ and total p3 and leads to an accumulation of their respective 12 kDa and 10 kDa APP precursor fragments in treated cells. These data suggest that the compound directly inhibits at least some form of γ-secretase although no data are provided as to what specific form of Aβ and p3 are affected.
Despite the progress which has been made in understanding the underlying mechanisms of AD, there remains a need for assays to identify candidate compounds for preventing or treating the disease.
SUMMARY OF THE INVENTION
According to current theory, the processing of APP is believed to involve several specific cleavages by proteases. The enzyme that cleaves APP between amino acids 671/672 (referring to the βAPP770 isoform) is called β-secretase. The enzyme that cleaves between amino acids 687/688 of APP (16/17 of Aβ) is called α-secretase. Until now it was believed that cleavage of APP that yielded Aβ(40) and Aβ(42) was carried out by a single enzyme called γ-secretase. However, we have discovered that a compound can inhibit the production of Aβ(40) but not Aβ(42). In particular, we have discovered that compounds, thought to inhibit the production Aβ in general, actually inhibit production of Aβ(40) but not Aβ(42). This indicates that multiple γ-secretase mechanisms are at work which can be pharmacologically dissociated.
Because Aβ(42) is the major component of β-amyloid plaques and initiates amyloid plaque formation in AD patients, it is important to have tools to screen compounds to identify those that specifically inhibit the production of Aβ(42) and Aβ(40), either simultaneously or separately. The current invention provides such assays.
This invention provides methods for determining whether a compound alters the amount of at least one Aβ(x-≧41) peptide produced by a cell. The methods involve administering the compound to a culture comprising the cell; measuring the amount of the Aβ(x-≧41) peptide, specifically, in a sample from the culture; and determining whether the measured amount is different than the amount expected in a sample from a culture comprising the cell to which no compound has been administered. A difference between the measured amount and the expected amount indicates that the compound alters the amount of an Aβ(x-≧41) peptide produced by the cell.
In another aspect, this invention provides methods for determining whether a compound alters the amount of at least one Aβ(x-≧41) peptide produced by a cell and alters the amount of either total Aβ or at least one Aβ(x-≦40) peptide produced by the cell. The methods involve administering the compound to a culture comprising the cell; measuring the amount of the Aβ(x-≧41) peptide, specifically, in a sample from the culture; measuring the amount of total Aβ or the Aβ(x-≦40) peptide, specifically, in a sample from the culture; and determining whether the measured amounts are different than the amounts expected in a sample from a culture comprising the cell to which no compound has been administered. Differences between the measured amounts and the expected amounts indicate that the compound alters the amount of the Aβ(x-≧41) peptide by a cell and/or the amount of total Aβ or the Aβ(x-≦40) peptide by the cell.
In one embodiment, the amount of the Aβ peptides are measured by immunoassay and, in particular, sandwich immunoassay comprising capture binding substances bound to a solid phase and a labeled detection binding substance.
In sandwich assays for determining the amount of at least one Aβ(x-≧41) peptide, the capture antibody preferably is specific for Aβ(x-≧41) peptides, e.g., raised against peptide NH2-Cys-NH—CH2—(CH2)5—CO-GLMVGGVVIA-COOH (SEQ ID NO: 4). The detection binding substance in this assay can be an antibody specific for Aβ peptides whose amino-terminal amino acid is no. 1 of Aβ, or can be specific for an epitope within the junction region of Aβ. In another embodiment the capture binding substance for measuring the amount of at least one of Aβ(x-≧41) peptide is specific for an epitope within the junction region of Aβ and the detection binding substance is an antibody specific for Aβ(x-≧41).
In sandwich assays for determining the amount of at least one Aβ(x-≦40) peptide, the capture binding substance preferably is an antibody specific for Aβ(x-≦40) peptides, e.g., raised against the peptide NH2-Cys-NH—CH2—(CH2)5—CO-GLMVGGVV-COOH (SEQ ID NO: 5). The labeled detection binding substance can be an antibody specific for the Aβ peptides whose amino-terminal amino acid is no. 1 of Aβ or an antibody specific for an epitope within the junction region of Aβ. In another embodiment the capture binding substance for measuring the amount of at least one of Aβ(x-≦40) peptide is specific for an epitope within the junction region of Aβ and the detection binding substance is an antibody specific for Aβ(x-≦40).
In sandwich assays for determining the amount of total Aβ, the capture binding substance preferably is an antibody specific for an epitope within the junction region of Aβ. The detection binding substance preferably is specific for Aβ peptides whose amino-terminal amino acid is no. 1 of Aβ.
In another embodiment of an immunoassay, the step of measuring the amount of the Aβ(x-≧41) peptide, total Aβ or the Aβ(x-≦40) peptide in a sample from the culture comprises: pulsing the culture with a radioactive label for protein; chasing the culture without a radioactive label; administering the compound to the cell during the chase period; contacting a sample from the culture with a binding substance specific for Aβ(x-≧41) peptides; contacting a sample from the culture with a binding substance specific for total Aβ or Aβ(x-≦40) peptide; and determining the amount of radioactive label attached to the binding substances.
In other embodiments of the methods the culture comprises primary human neurons, primary neurons from a transgenic PDAPP mouse (i.e., a transgenic mouse whose cells harbor a PDAPP construct), a 293 human kidney cell line, a human neuroglioma cell line, a human HeLa cell line, a primary endothelial cell line, a primary human fibroblast line, a primary lymphoblast line, human mixed brain cells, or a Chinese hamster ovary (CHO) cell line. In one embodiment the cell is a host cell transfected with a recombinant expression vector encoding a human APP, e.g., a Hardy mutation such as V717F or the Swedish mutant; causing the cell to overproduce Aβ(x-≧41) peptides. In another aspect, the methods further comprise the step of determining whether the compound is toxic to the cell.
In another aspect this invention provides kits for specifically detecting at least one Aβ(x-≧41) peptide and at least one Aβ(x-≦40) peptide in a sample. The kits include a binding substance specific for Aβ(x-≧41) peptides; and a binding substance specific for Aβ(x-≦40) peptides.
In another aspect this invention provides kits for specifically detecting at least one Aβ(x-≧41) peptide and either total Aβ or at least one Aβ(x-≦40) peptide in a sample in a sandwich immunoassay. The kits include at least two different binding substances for measuring the amount of Aβ(x-≧41) peptide; and at least two different binding substances for measuring the amount of total Aβ or Aβ(x-≦40) peptides.
In another aspect this invention provides methods for determining whether a compound alters the amount of at least one Aβ(x-≧41) peptide produced by a non-human mammal and alters the amount of either total Aβ or at least one Aβ(x-≦40) peptide produced in the non-human mammal. The methods involve measuring a first amount of the Aβ(x-≧41) peptide in a sample from a non-human animal used as a model of Alzheimer's disease; measuring a first amount of total Aβ or the Aβ(x-≦40) peptide in a sample from the non-human animal; administering the compound to the non-human animal; measuring a second amount of the Aβ(x-≧41) peptide in a sample from the non-human animal; measuring a second amount of total Aβ or the Aβ(x-≦40) peptide in a sample from the non-human animal; and comparing the first amounts with the second amounts. The comparison indicates whether the compound increases, decreases, or leaves unchanged the amount of the Aβ(x-≧41) peptide and increases, decreases, or leaves unchanged the amount of the Aβ(x-≦40) peptide. In certain embodiments the non-human animal is a rodent, in particular, a mouse. The non-human animal can harbor a copy of an expressible transgene sequence which encodes a Hardy mutation (e.g., V717F) or the Swedish mutation of human β-amyloid precursor protein (APP).
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1. Shows standard curves of Aβ(1-40) and Aβ(1-42) with 2G3 as a capture antibody.
FIG. 2. Shows standard curves of Aβ(1-40) and Aβ(1-42) with 21F12 as a capture antibody.
FIG. 3. Shows that compound MDL 28170 influences βAPP metabolism. (0) untreated K695sw cells; (200) K695sw cells treated with 200 μM MDL 28170 during the chase period. Lanes 1,2: aliquot of total chase media run directly on the gel. Lanes 3,4: antibody 1736 immunoprecipitation of chase media. Lanes 5,6: antibody 192sw immunoprecipitation of chase media. Note that β-cut APPs (lanes 5,6) runs slightly below α-cut APPs (lanes 3,4), as expected. Lanes 7,8: antibody C7 immunoprecipitation of cell lysates. Lanes 9,10: antibody 1282 immunoprecipitation of cell lysates.
FIGS. 4A-4B. Differential inhibition of Aβ(42) and Aβ(40) formation. (A) Labeled K695sw cells were chased with the indicated concentrations of MDL 28170, and the chase media were precipitated with 21F12 (upper panel) followed by antibody 1282 (lower panel). (B) Quantitation of the effect of 200 μM MDL 28170 on Aβ and p3 in the chase media by phosphorimaging. The bars show the relative pixel number compared to an untreated control (set at 100%). The decreases in Aβtotal and p3total relative to an untreated control were significant (*) (two-tailed t-test, n=4, p<0.001), whereas the decreases in Aβ(42) and p3(42) upon treatment with MDL 28170 did not reach significance. The difference in reduction of Aβ(42) level vs. total Aβ level is significant (two-tailed t-test, n=4, p<0.01). The difference in inhibition of p3(42) vs. total p3 is also significant (two-tailed t-test, n=4, p<0.05).
FIGS. 5A-5F. Differential inhibition of Aβ production by K695sw cells under a variety of conditions. (A) treatment with 1 μM PDBu (P) decreases both Aβ(42) and Aβtotal. Upper panel: antibody 21F12 precipitation, lower panel: subsequent antibody 1282 precipitation. (B-E) K695sw cells were chased with (M) or without (0) 200 μM MDL 28170, and the chase media were immunoprecipitated (B) first with antibody BC05 (upper panel) and then with antibody 1282 (lower panel); (C) first with antibody C42 (upper panel) and then with antibody 1282 (lower panel); (D) first with antibody 2G3 (upper panel) and then with antibody 21F12 (lower panel); (E) after splitting the medium into two aliquots, either with antibody 21F12 (upper panel) or antibody 1282 (lower panel). (F) cells were labeled for 3 h in the presence of 100 μM MDL 28170, and the medium was immunoprecipitated first with antibody 21F12 (upper panel) and then with antibody 1282 (lower panel).
FIGS. 6A-6C. Differential inhibition of Aβ production in different cell types. 35S-methionine labeled cells were chased with the indicated concentrations of MDL 28170 and precipitated with antibody 21F12 (upper panel) followed by antibody 1282 (lower panel). (A) V717F cells. The relatively low APP expression leads to a faint Aβ42 band. (B) CHO695 cells. Note that in CHO cells the p3 brands migrate as doublets, as described (Koo and Squazzo, (1994) “Evidence that production and release of amyloid β-protein involves the endocytic pathway.” J. Biol. Chem. 269:17386-17389). (C) SKN695 cells. While Aβ(42) and p3(42) are slightly increased at 200 μM MDL 28170, Aβtotal and p3total are decreased.
FIG. 7. Shows a summary sketch of some of the known proteolytic processing pathways of APP.
FIG. 8A. Shows a schematic of a combination cDNA/genomic coding sequence allowing alternative splicing of the KPI and OX-2 exons.
FIG. 8B. Shows a schematic of a combination cDNA/genomic coding sequence bearing, a mutation at position 717 and allowing alternative splicing of the KPI and OX-2 exons.
FIG. 9. Diagram of the intermediate constructs used to construct the APP splicing cassette and the PDAPP vector.
FIGS. 10A-H. The DNA sequence (SEQ ID NO: 2) and the deduced amino acid sequence (SEQ ID NO: 3) of APP770.
FIG. 11. Diagram, of the genomic region of APP present in the PDAPP construct. The sizes of original introns 6, 7 and 8, as well as the sizes of the final introns are indicated in the diagram. The locations of the deletions in introns 6 and 8 present in the PDAPP construct also are indicated.
FIG. 12. Schematic map of the PDAPP vector, a combination cDNA/genomic APP construct.
FIGS. 13A-13O depict the percent inhibition of Aβ by several compounds using the brain cell culture method described in Example VIII. The charts show inhibition of total Aβ (“% Aβ inhibition”), inhibition of Aβ(42) (“% 1-42 inhibition”) and inhibition of the metabolism of MTT (“% MTT inhibition”) (greater inhibition indicates greater cytotoxicity).
DETAILED DESCRIPTION OF THE INVENTION
I. Definitions
The term “binding substance” refers to a polypeptide substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof, which specifically bind and recognize an analyte (antigen). The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as the myriad immunoglobulin variable region genes. Antibodies exist, e.g., as intact immunoglobulins or as a number of well characterized fragments produced by digestion with various peptidases. This includes, e.g., Fab′ and F(ab)′2 fragments. The term “binding substance,” as used herein, also includes antibody fragments either produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA methodologies.
The term “immunoassay” is an assay that utilizes a binding substance to specifically bind an analyte. The immunoassay is characterized by the use of specific binding properties of a particular antibody to isolate, target, and/or quantify the analyte.
A binding substance “specifically binds to” or “is specifically immunoreactive with” a protein when the binding substance functions in a binding reaction which is determinative of the presence of the protein in the presence of a heterogeneous population of proteins and other biologics. Thus, under designated immunoassay conditions, the specified binding substances bind preferentially to a particular protein and do not bind in a significant amount to other proteins present in the sample. Specific binding to a protein under such conditions requires an antibody that is selected for its specificity for a particular protein. A variety of immunoassay formats may be used to select binding substances specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York, for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity.
A “label” is a composition detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means. For example, useful labels include 32P, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, dioxigenin, or haptens and proteins for which antisera or monoclonal antibodies are available. Binding substances can be made detectible, e.g., by incorporating a radio-label into the peptide, and used to detect antibodies specifically reactive with the peptide. A label often generates a measurable signal, such as radioactivity, fluorescent light or enzyme activity, which can be used to quantitate the amount of bound label.
II. In Vitro Screening
Alzheimer's disease is characterized by the initial deposition of Aβ(x-≧41) in the form of amyloid plaques in the brain. Therefore, effective treatments for AD are expected to decrease the production of these peptides, whereas agents that hasten progress of the disease are expected to increase production of the peptide. Prior screening methods looked for compounds that decreased total Aβ. However, since Aβ(x-≧41) peptides are a small fraction of total Aβ, those assays could not determine whether the compound specifically inhibited Aβ(x-≧41) peptides. As results described herein indicate, compounds can alter the production of Aβ(x-≦40) but not Aβ(x-≧41). Because Aβ(42) is the major constituent of neuritic plaques, it is useful to identify compounds that specifically inhibit the production of Aβ(x-≧41) peptides, either in addition to, or instead of Aβ(x-≦40) peptides. Accordingly, this invention provides methods for screening compounds that specifically elevate or decrease the production of the amount of Aβ(x-≧41) by a cell and compounds that elevate or decrease production of both Aβ(x-≧41) and Aβ(x-≦40) (e.g., total Aβ), or of one or the other of these peptides. Compounds that decrease production of Aβ(x-≧41) are candidates for use in treating the disease, while compounds that increase its production may hasten the disease and are to be avoided by humans.
Screening methods of this invention for determining whether a test compound specifically alters the amount of Aβ(x-≧41) produced by a cell involve administering the compound to the cell, usually in culture, measuring the amount of Aβ(x-≧41) specifically produced by the cell, and determining whether this amount is greater than, less than or the same as an amount the cell is expected to produce in the absence of the compound. If the amounts are different, then the compound affects the production of Aβ(x-≧41) by the cell. This amount can be measured, for example, in a sample from the culture, such as medium conditioned by the cell in culture, or in extracts derived from cells harvested from the culture.
The expected amount generally will be a control amount determined by measuring Aβ(x-≧41) produced by the cell in the absence of the compound. However, one also may determine the expected amount by extrapolation; measuring the amount of Aβ(x-≧41) produced upon administration of different amounts of the compound to the cell, and using these figures to calculate the expected amount. In certain instances measuring a control amount for the purposes of comparison may not be necessary because the effect of the compound on Aβ(x-≧41) production is clear. For example, a compound may render Aβ(x-≧41) undetectable in a cell that normally produces detectable amounts, indicating that the compound decreases Aβ(x-≧41) production from the amount expected in its absence.
In another aspect, this invention provides screening methods for determining whether a compound alters the production of Aβ(x-≧41) by a cell to a different degree than it alters the production of total Aβ or Aβ(x-≦40) by the cell. These methods are useful for determining whether a compound alters the production of Aβ(x-≧41) in addition to total Aβ, or alters the production of one or the other of Aβ(x-≧41) and Aβ(x-≦40). The methods involve administering the compound to the cell (usually in culture). Then, the degree to which the compound alters the production of Aβ(x-≧41), specifically, by the cell is determined. The degree to which the compound alters the production of total Aβ or Aβ(x-≦40) by the cell also is determined. Then, the degrees are compared. The comparison indicates whether the compound alters the production of Aβ(x-≧41) instead of or in addition to Aβ(x-≦40).
Determining the degree to which a compound alters the production of one or the other peptide generally involves measuring the specific amount of the peptide in a sample from the culture; and comparing it with the amounts expected in a sample from a culture comprising the cell to which no compound has been administered.
III. Amyloid-β Peptide and Related Proteins and Peptides
Various cellular processing pathways for APP are presented in FIG. 7. The terms “amyloid-β peptide,” “Aβ” or “βAP” as used herein refer to an approximately 4.2 kD protein which, in the brains of AD, Down's Syndrome, HCHWA-D and some normal aged subjects, forms the subunit of the amyloid filaments comprising the senile (amyloid) plaques and the amyloid deposits in small cerebral and meningeal blood vessels (amyloid angiopathy). Aβ can occur in a filamentous polymeric form (in this form, it exhibits the Congo-red and thioflavin-S dye-binding characteristics of amyloid described in connection therewith). Aβ can also occur in a non-filamentous form (“preamyloid” or “amorphous” or “diffuse” deposits) in brain tissue, in which form no birefringent staining by Congo red occurs. A portion of this protein in the insoluble form obtained from meningeal blood vessels is described in U.S. Pat. No. 4,666,829.
Aβ is an approximately 39-43 amino acid fragment of a large membrane-spanning glycoprotein, referred to as the β-amyloid precursor protein (APP), encoded by a gene on the long arm of human chromosome 21. Forms of Aβ longer than 43 amino acids are also contemplated herein. Aβ is further characterized by its relative mobility in SDS-polyacrylamide gel electrophoresis or in high performance liquid chromatography (HPLC). A sequence for a 43-amino acid-version of Aβ is:
(SEQ ID NO:1)
1
Asp Ala Glu Phe Arg His Asp Ser Gly Tyr
11
Glu Val His His Gln Lys Leu Val Phe Phe
21
Ala Glu Asp Val Gly Ser Asn Lys Gly Ala
31
Ile Ile Gly Leu Met Val Gly Gly Val Val
41
Ile Ala Thr.
As used herein, Aβ also refers to related polymorphic forms of Aβ, including those that result from mutations in the Aβ region of the APP gene.
The term “Aβ fragment” as used herein refers to fragments and degradation products of Aβ which are generated at low concentrations by mammalian cells. Particular Aβ fragments have a molecular weight of approximately 3 kD and are presently believed to include peptides with, for example, amino acid residues 3-34, 6-27, 6-34, 6-35, 6-42, 11-34, 11-40, 11-43, 12-43, 17-40 and 17-42 of Aβ (Vigo-Pelfrey et al. (1993) J. Neurochem. 61:1965-1968).
As used herein, the term “Aβ(x-≧41)” refers to Aβ or an Aβ fragment whose amino-terminus begins at amino acid number 1 of Aβ or thereafter (i.e., which is amino-terminally truncated), and whose carboxy-terminus extends beyond amino acid number 40. These peptides and fragments comprise a heterogenous group. For example, Aβ(6-42), Aβ(11-43) and Aβ(12-43) all have been found in the CSF. However, this list is not meant to be exclusive. Other peptides from among the group are presumed to exist in the culture media of cells expressing APP and are detectable with the methods described herein. As used herein the term “Aβ(42)” refers to Aβ or an Aβ fragment whose C-terminal amino acid is # 42 of Aβ.
As used herein, the term “Aβ(x-≦40)” refers to Aβ or an Aβ fragment whose amino-terminus begins at amino acid number 1 of Aβ or which is amino-terminally truncated, and whose carboxy-terminus extends no further than amino acid number 40. These peptides and fragments also comprise a heterogenous group. The term “Aβ(40)” refers to Aβ or an Aβ fragment whose C-terminal amino acid is #40 of Aβ.
As used herein, the term “p3” refers to a peptide whose amino acid sequence is substantially similar to Aβ, but whose amino-terminal amino acid begins at amino acid 17 of Aβ. The term “p3 fragment” as used herein refers to fragments and degradation products of p3. Whereas p3 is produced through a different processing pathway than Aβ, for the purposes of the detection methods of this invention, p3 and p3 fragments are considered to be a subset of Aβ peptides, because certain detection techniques that recognize Aβ solely from the carboxy-terminus generally also will recognize p3. Also it is shown that the same apparent mechanisms generate the p3 and Aβ carboxy-termini.
The term “Aβ junction region” as used herein refers to a region of Aβ which is centered at the site between amino acid residues 16 and 17 (Lys16 and Leu17), which is a principal target for proteolytic processing of APP. Such processing, referred to as “α-secretory” processing, results in a variety of APP fragments which may, for example, terminate at amino acid 16 of Aβ and which, therefore, are potentially immunologically cross-reactive with antibodies to the intact Aβ molecule which are to be used in the methods of the present invention. Antibodies raised against a synthetic peptide including amino acid residues 13-28 have been found to display the requisite specificity for the junction region.
The term “amyloid-β precursor protein” (APP) as used herein is defined as a polypeptide that is encoded by a gene of the same name localized in humans on the long arm of chromosome 21 and that includes the Aβ region within the carboxyl third of its coding region. APP is a glycosylated, single-membrane-spanning protein expressed in a wide variety of cells in many mammalian tissues. Examples of specific isotypes of APP which are currently known to exist in humans are the 695-amino acid polypeptide described by Kang et al. (1987) Nature 325:733-736; the 751-amino acid polypeptide described by Ponte et al. (1988) Nature 331:525-527 (1988) and Tanzi et al. (1988) Nature 331:528-530; and the 770-amino acid polypeptide described by Kitaguchi et al. (1988) Nature 331:530-532. Examples of specific variants of APP include point mutations which can differ in both position and resultant neuropathological phenotype (for review of known variant mutations see Hardy (1992) Nature Genet. 1:233-234).
The term “Aβ-related condition” as used herein is defined as including Alzheimer's disease (which includes familial Alzheimer's disease), Down's Syndrome, HCHWA-D, and advanced aging of the brain.
IV. Cells Expressing Aβ(x-≧41)
Because it is a therapeutic goal to discover compounds that inhibit the production of Aβ(x-≧41), the test cells used in the methods of this invention generally are ones that are able to secrete Aβ(x-≧41).
In vitro monitoring of Aβ(x-≧41) levels in conditioned medium from a suitable cell culture may be used for drug screening. By growing cells under conditions which result in the secretion of Aβ(x-≧41) into the culture medium, and exposing the cells to test compounds, the effect of these test compounds on Aβ(x-≧41) secretion can be observed.
Suitable cell lines include human and animal cell lines, such as, preferably, primary human neurons, and primary neurons from transgenic mice harboring human APP genes, e.g., cells from a transgenic PDAPP animal (e.g., mouse), as well as a 293 human kidney cell line, a human neuroglioma cell line, a human HeLa cell line, a primary endothelial cell line (e.g., HUVEC cells), a primary human fibroblast line or a primary lymphoblast line (including endogenous cells derived from patients with APP mutations), a primary human mixed brain cell culture (including neurons, astrocytes and neuroglia), or a Chinese hamster ovary (CHO) cell line. Particularly useful are cells stably transfected with APP695 carrying the mutation V717I (valine to isoleucine at position 717 in the APP770 numbering system). Cell lines which preferentially increase the levels or ratios of Aβ(x-≧41) would be particularly useful in the methods of invention. Useful mutants at position 717 (the Hardy mutation) include V717F, V717I or V717G.
Preferred for use in drug screening methods according to the present invention are cell lines capable of expressing APP variants which overproduce Aβ. By “overproduce,” it is meant that the amount of Aβ produced from the variant APP will be at least about one-and-a-half times and preferably at least two or five times greater than the amount produced from any or all of the normal APP isoforms, e.g., the 695, 751, and 770 amino acid isoforms which have been previously described. Particularly preferred are APP variants having one or several amino acid substitutions directly amino-terminal of the Aβ cleavage site. For example, K293 cells which express an APP695 DNA bearing a double mutation (Lys595->Asn595 and Met596->Leu596 (695 numbering system)) found in a Swedish FAD family produce approximately five-to-eight-fold more Aβ than cells expressing normal APP (Citron et al. (1992) Nature, 360:672-674). The mutation at residue 596 appears to be principally responsible for the increase.
V. Expression Vectors for APP
Host cells transfected with a recombinant expression vector that encodes APP also are useful as cells in the screening methods of this invention. A plasmid that carries sequences encoding APP is pCMV695 (Selkoe et al. (1988) Proc. Natl. Acad. Sci USA 85:7341-7345).
Nucleic acids encoding APP can be obtained by methods known in the art. For example, a nucleic acid encoding an APP can be isolated by polymerase chain reaction of cDNA or genomic DNA from a human brain cDNA library or a human genomic library using primers based on the DNA sequence of APP. PCR methods are described in, for example, U.S. Pat. No. 4,683,195; Mullis et al. (1987) Cold Spring Harbor Symp. Quant. Biol. 51:263; and Erlich, ed., PCR Technology, (Stockton Press, NY, 1989).
Mutant versions of APP, such as the Swedish mutation, can be made by site-specific mutagenesis of other nucleic acids encoding APP, or by random mutagenesis caused by increasing the error rate of PCR of the original polynucleotide with 0.1 mM MnC1 2 and unbalanced nucleotide concentrations.
The construction of expression vectors and the expression of genes in transfected cells involves the use of molecular cloning techniques also well known in the art. Sambrook et al., Molecular Cloning—A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., (1989) and Current Protocols in Molecular Biology, F. M. Ausubel et al., eds., (Current Protocols, a joint venture between Greene Publishing Associates, Inc. and John Wiley & Sons, Inc.).
Nucleic acids used to transfect cells with sequences coding for expression of the polypeptide of interest generally will be in the form of an expression vector that includes expression control sequences operatively linked to a nucleotide sequence coding for expression of the polypeptide. As used herein, the term nucleotide sequence “coding for expression of” a polypeptide refers to a sequence that, upon transcription and translation of mRNA, produces the polypeptide. As any person skilled in the art recognizes, this includes all degenerate nucleic acid sequences encoding the same amino acid sequence. This can include sequences containing, e.g., introns. As used herein, the term “expression control sequences” refers to nucleic acid sequences that regulate the expression of a nucleic acid sequence to which it is operatively linked. Expression control sequences are “operatively linked” to a nucleic acid sequence when the expression control sequences control and regulate the transcription and, as appropriate, translation of the nucleic acid sequence. Thus, expression control sequences can include appropriate promoters, enhancers, transcription terminators, a start codon (i.e., ATG) in front of a protein-encoding gene, splicing signals for introns, maintenance of the correct reading frame of that gene to permit proper translation of the mRNA, and stop codons.
The recombinant nucleic acid can be incorporated into an expression vector comprising expression control sequences operatively linked to the recombinant nucleic acid. The expression vector can be adapted for function in prokaryotes or eukaryotes by inclusion of appropriate promoters, replication sequences, markers, etc.
The expression vector can be transfected into a host cell for expression of the recombinant nucleic acid. Host cells can be selected for high levels of expression in order to purify the protein. The host cell can be a prokaryotic or eukaryotic cell selected to study the activity of an enzyme produced by the cell. The cell can be, e.g., a cultured cell or a cell in vivo.
Transfected host cells useful in this invention include human kidney 293 cell lines such as K695sw, and K6957171, glioma cell lines such as HS695, and neuroblastoma cell lines such as SKN695, described in the Experimental section.
VI. Measuring Aβ(x-≧41)
Aβ peptides can be detected by any method known in the art. Preferably, the method involves an immunoassay employing binding substances specific for the peptides. Optionally, one can detect Aβ peptides by determining their size, e.g., by HPLC or by mass spectrometry.
A. Binding Substances
One step of the screening methods of this invention involves measuring the amount of at least one Aβ(x-≧41) peptide, specifically, in a sample. Measuring Aβ(x-≧41) peptides specifically means measuring Aβ(x-≧41) peptides so as to distinguish that molecule from shorter species of Aβ, i.e., those species whose carboxy-terminus extends no further than amino acid # 40 of Aβ.
Specific measurement of Aβ(x-≧41) preferably is performed by the use of binding substances that specifically recognize Aβ(x-≧41) peptides, e.g., binding substances that recognize amino acids of Aβ beyond amino acid # 40.
Another method of this invention involves screening compounds to determine their ability to alter the production of both Aβ(x-≧41) peptides and total Aβ or Aβ(x-≦40) peptides. Such methods can involve the use of binding substances that can distinguish Aβ(x-≦40) peptides from longer species of Aβ, such as Aβ(x-≧41) peptides.
B. Immunoassays
The use of immunological detection techniques, i.e., immunoassays employing binding substances, is preferred. Particularly suitable detection techniques include ELISA, Western blotting, radioimmunoassay, and the like. Suitable. immunological methods employing a single antibody are also contemplated, for example, radioimmunoassay using an antibody specific for ≧41 forms of Aβ, or single antibody ELISA methods. It will be clear that the particular forms of Aβ detected by such methods depend upon the particular binding substances employed. For example, binding substances directed to the junction region may detect Aβ(x-≧41) peptides whose amino termini do not extend to amino acid no. 1 of Aβ. Also, binding substances directed to the carboxy-terminal end of Aβ(x-≧41) may detect peptides ending at amino acids 41, 42 or 43. Therefore, determining the specificity of the binding substances will assist in determining exactly which Aβ(x-≧41) peptides the assay is detecting.
In one embodiment, the method to detect Aβ(x-≧41) peptides is an immunoassay involving two antibodies. One antibody is specific for an epitope containing amino acids beyond number 40 in Aβ, and another antibody is capable of distinguishing Aβ and Aβ fragments from other APP fragments which might be found in the sample. In particular, it has been found that antibodies which are monospecific for the junction region of Aβ are capable of distinguishing Aβ from other APP fragments. The junction region of Aβ is centered at amino acid residues 16 and 17, typically spanning amino acid residues ˜13 to ˜28. Such “junction-recognizing” antibodies may be prepared using synthetic peptides having that sequence as an immunogen.
A preferred immunoassay technique is a two-site or “sandwich” assay. This assay involves a capture binding substance, usually bound to a solid phase, and a labelled detection binding substance. In this method, Aβ(x-≧41) peptides are captured from the sample using a first binding substance specific for Aβ(x-≧41) peptides (usually bound to a solid phase). The capture of Aβ(x-≧41) peptides is detected using a labeled second binding substance specific for Aβ. Labeled binding substances include, for example, those directed to the junction region (amino acids ˜13 to ˜28) or binding substances specific for amino-terminal amino acids (1-5 or 1-12).
Particular methods for preparing such antibodies and utilizing such antibodies in an exemplary ELISA are set forth in the Experimental section hereinafter and in related U.S. patent application Ser. No. 07/965,972, supra. A sandwich assay using an antibody against the junction region can be used to specifically measure Aβ and Aβ fragments whose amino-terminus begins before amino acid 13 of Aβ. Such assays do not recognize p3 or p3 fragments, since those peptides begin at amino acid # 17 of Aβ.
Antibodies specific for Aβ(x-≧41), i.e., which do not cross react with Aβ(≦40), are particularly useful in the methods of this invention. These antibodies can be made by immunizing animals with synthetic peptides that include amino acids beyond number 40 of Aβ. For example, the synthetic peptide can include amino acids 33-42. A specific example of the production of such an antibody is provided in the Experimental section.
The particular peptides measured from among the group of all Aβ(x-≧41) depends on the particular measuring method used. In the case of using binding substances, such as antibodies, the binding substance can be directed to one or more from among the group of peptides. For example, an antibody raised against amino acids 33-42 of Aβ that does not cross react with Aβ(1-40) will bind to Aβ(x-42). It also may bind to Aβ(x-41) and Aβ(x-43). According to one embodiment of, the invention, the method involves determining the amount of Aβ(x-≧41) having at least amino acids 13-41 of Aβ. These species can be measured using a sandwich assay employing antibodies that recognize the junction region (amino acids 13-26) and antibodies produced by immunization with a hapten having Aβ amino acids 33-42. Total Aβ can be measured using a capture antibody to the junction region (e.g., the 266 antibody, described herein) and a reporter antibody that should detect virtually all the Aβ peptides and Aβ fragments, e.g., an antibody raised against amino acids 1-12 of Aβ.
C. Pulse-Chase Assays
Another method of measuring the amount of Aβ(x-≧41) in a sample involves pulse-chase procedures. In these methods, the culture is pulsed with a radioactive label for protein, e.g., a radioactive amino acid such as 35S methionine. Then the culture is chased without the label. Then the compound is administered to the cells. Then the cells are contacted with a binding substance specific for Aβ(x-≧41) and the amount of radioactive label attached to the binding substance is determined.
D. Preparation of Antibodies
Antibodies specific for Aβ can be prepared, e.g., by immunizing an animal with a peptide whose amino acid sequence corresponds with amino acids ˜13 to ˜28 of Aβ. Antibodies specific for Aβ(x-≧41) can be prepared, e.g., by immunizing an animal with a peptide whose amino acid sequence corresponds with amino acids 33-42 of Aβ. Antibodies specific for Aβ(x-≦40) can be prepared, e.g., by immunizing an animal with a peptide whose amino acid sequence corresponds with amino acids 33-40 or 28-40 of Aβ. Antibodies against the junction region are useful for detecting total Aβ.
Synthetic polypeptide haptens may be produced by the well-known Merrifield solid-phase synthesis technique in which amino acids are sequentially added to a growing chain (Merrifield (1963) J. Am. Chem. Soc. 85:2149-2156). Suitable peptide haptens will usually comprise at least five contiguous residues within Aβ and may include more than six residues. The amino acid sequences may be based on the sequence of Aβ set forth above.
Once a sufficient quantity of polypeptide hapten has been obtained, it may be conjugated to a suitable immunogenic carrier, such as serum albumin, keyhole limpet hemocyanin, or other suitable protein carriers, as generally described in Hudson and Hay, Practical Immunology, Blackwell Scientific Publications, Oxford, Chapter 1.3, 1980. An exemplary immunogenic carrier utilized in the examples provided below is α-CD3ε antibody (Boehringer-Mannheim, Clone No. 145-2C11).
Antibodies specific for the desired epitope may be produced by in vitro or in vivo techniques. In vitro techniques involve exposure of lymphocytes to the immunogens, while in vivo techniques require the injection of the immunogens into a suitable vertebrate host. Suitable vertebrate hosts are non-human, including mice, rats, rabbits, sheep, goats, and the like. Immunogens are injected into the animal according to a predetermined schedule, and the animals are periodically bled, with successive bleeds having improved titer and specificity. The injections may be made intramuscularly, intraperitoneally, subcutaneously, or the like, and an adjuvant, such as incomplete Freund's adjuvant, may be employed.
If desired, monoclonal antibodies can be obtained by preparing immortalized cell lines capable of producing antibodies having desired specificity. Such immortalized cell lines may be produced in a variety of ways. Conveniently, a small vertebrate, such as a mouse, is hyperimmunized with the desired immunogen by the method just described. The vertebrate is then killed, usually several days after the final immunization, the spleen cells removed, and the spleen cells immortalized. The manner of immortalization is not critical. Presently, the most common technique is fusion with a myeloma cell fusion partner, as first described by Kohler and Milstein (1975) Nature 256:495-497. Other techniques including EBV transformation, transformation with bare DNA, e.g., oncogenes, retroviruses, etc., or any other method which provides for stable maintenance of the cell line and production of monoclonal antibodies. Specific techniques for preparing monoclonal antibodies are described in Antibodies: A Laboratory Manual, Harlow and Lane, eds., Cold Spring Harbor Laboratory, 1988.
In addition to monoclonal antibodies and polyclonal antibodies (antisera), the detection techniques of the present invention will also be able to use antibody fragments, such as F(ab), Fv, VL, VH, and other fragments. In the use of polyclonal antibodies, however, it may be necessary to adsorb the anti-sera against the target epitopes in order to produce a monospecific antibody population. It will also be possible to employ recombinantly produced antibodies (immunoglobulins) and variations thereof as now well described in the patent and scientific literature. See, for example, EPO 8430268.0; EPO 85102665.8; EPO 85305604.2; PCT/GB 85/00392; EPO 85115311.4; PCT/US86/002269; and Japanese application 85239543, the disclosures of which are incorporated herein by reference. It would also be possible to prepare other recombinant proteins which would mimic the binding specificity of antibodies prepared as just described.
VII. Kits
This invention also provides kits for performing assays of the invention. The kits include means for detecting specifically Aβ(x-≧41) and means for detecting specifically Aβ(x-≦40). The means can include any means known or described above, e.g., binding substances.
In one embodiment, the kit includes a binding substance specific for Aβ(x-≧41) (i.e., that does not cross react with Aβ(≦40)), and a binding substance specific for Aβ(≦40) (i.e., that does not cross react with Aβ(x-≧41)). Such kits are useful, e.g., in pulse-chase assays.
In another embodiment, the kit is useful for immunoassays including two antibodies for each antigen. For example, the kit can further comprise a binding substance specific for the junction region of Aβ. Such antibodies are useful for the capture or detection of both Aβ(x-≧41) and Aβ(≦40). In one kit useful for a sandwich ELISA, the binding substance specific for the junction region is bound to a solid phase, and the binding substances specific for Aβ(x-≧41) and Aβ(≦40) are detectably labeled.
The detectable labels can be any known and used in the art including, e.g., a biotinylation label, a radioactive label, a light scattering label, an enzymatic label, a fluorescent label and the like. When the label is enzymatic, the kit can further comprise a substrate for the enzyme.
VIII. Test Compounds
The test compounds can be any molecule, compound, or other substance which can be added to the cell culture or administered to the test animal without substantially interfering with cell or animal viability. Suitable test compounds may be small molecules (i.e., molecules whose molecular mass is no more than 1000 Daltons), biological polymers, such as polypeptides, polysaccharides, polynucleotides, and the like. The test compounds will typically be administered to the culture medium at a concentration in the range from about 1 nM to 1 mM, usually from about 10 μM to 1 mM. The test compounds will typically be administered at a dosage of from 1 ng/kg to 100 mg/kg, usually from 10 μg/kg to 1 mg/kg.
Test compounds which are able to inhibit secretion or production of Aβ(x-≧41) are considered as candidates for further determinations of the ability to block β-amyloid production in animals and humans. Such compounds can be tested in in vivo studies, as described below. Inhibition of secretion or production indicates that cleavage of Aβ between amino acids 42/43 has likely been at least partly blocked, reducing the amount of Aβ(x-≧41) available for forming β-amyloid plaques.
IX. In Vivo Screening
Animal models currently are being used to study Alzheimer's disease. (See, e.g., International Patent Application WO 93/14200, U.S. patent application Ser. No. 08/143,697, filed Oct. 27, 1993, now U.S. Pat. No. 5,604,102, U.S. Pat. No. 5,387,742, and U.S. application Ser. No. 08/486,538, filed Jun. 7, 1995.) These models are useful for screening compounds that alter the production of Aβ(x-≧41) in the assays of this invention for their ability to affect the course of Alzheimer's disease, both to ameliorate and aggravate the condition. Transgenic mammalian models, more particularly, rodent models and in particular murine, hamster and guinea pig models, are suitable for this use.
A preferred non-human transgenic animal is one whose cells harbor a PDAPP construct. A PDAPP construct is a nucleic acid construct that comprises a mammalian promoter operatively linked to a cDNA-genomic DNA hybrid coding for the expression of APP. The cDNA-genomic DNA hybrid contains a cDNA sequence encoding APP770 or a cDNA sequence encoding APP770 with a naturally occurring mutation (e.g., a Hardy mutation or the Swedish mutation) substituted with genomic DNA sequences. The genomic DNA sequences consist of exon 6 and an amount of the adjacent downstream intron sufficient for splicing, the KI and OX-2 coding region and an amount of each of their upstream and downstream introns sufficient for splicing, and exon 9 and an amount of the adjacent upstream intron sufficient for splicing, substituted into the corresponding region of the cDNA sequence encoding APP770, or the cDNA encoding APP770 with a naturally occurring mutation. The construct is transcribed and differentially spliced in mammalian cells to form mRNA molecules that encode and that are translated into APP695, APP751 and APP770. In certain embodiments, the construct contains a PDGF-β promoter operatively linked with a hybrid sequence encoding an APP gene harboring a Hardy mutation (V717F), and the SV40 polyadenylation signal. One version of the PDAPP construct is presented in Example IX.
Another useful non-human animal model harbors a copy of an expressible transgene sequence which encodes the Swedish mutation of APP (asparagine595-leucine596). The sequence generally is expressed in cells which normally express the naturally-occurring endogenous APP gene (if present). Such transgenes typically comprise a Swedish mutation APP expression cassette, in which a linked promoter and, preferably, an enhancer drive expression of structural sequences encoding a heterologous APP polypeptide comprising the Swedish mutation. Aβ levels can be measured in any body fluid or tissue sample, for example, brain homogenate.
The transgenic animals that are usually produced by introducing the transgene or targeting construct into a fertilized egg or embryonic stem (ES) cell, typically by microinjection, electroporation, lipofection, or biolistics. The transgenic animals express the Swedish mutation APP gene of the transgene (or homologously recombined targeting construct), typically in brain tissue. Preferably, one or both endogenous APP alleles is inactivated and incapable of expressing the wild-type APP.
In all cases, it will be necessary to obtain a control value which is characteristic of the level of Aβ(x-≧41) and/or total Aβ or Aβ(x-≦40) production in the test animal in the absence of test compound(s). In cases where the animal is sacrificed, it will be necessary to base the value on an average or a typical value from other test animals which have been transgenically modified to express the Swedish mutant of human APP but which have not received the administration of any test compounds or any other substances expected to affect the level of production of Aβ(x-≧41) and/or total Aβ or Aβ(x-≦40). Once such control level is determined, test compounds can be administered to additional test animals, in which deviation from the average control value would indicate that the test compound had an effect on the γ-secretase activity in the animal. Test substances which are considered positive, i.e., likely to be beneficial in the treatment of Alzheimer's disease or other β-amyloid-related conditions, will be those which are able to reduce the level of Aβ(x-≧41) production, preferably by at least 20%, more preferably by at least 50%, and most preferably by at least 80%.
The following examples are offered by way of illustration, not by way of limitation.
EXPERIMENTAL
I. Antibody Preparation
A. Monoclonal Antibodies to the Aβ Junction Region
Monoclonal antibodies to the junction region of Aβ were prepared using a synthetic peptide spanning amino acid residues 13-30, except that AI, amino acids 30 and 31, were substituted with GC (the “junction peptide”). The junction peptide was conjugated to an immunogen (α-CD3ε antibody; Clone No. 145-2C11, Boehringer-Mannheim) using m-maleimidobenzoyl-N-hydroxysuccinimide ester (MHS) according to the manufacturer's (Pierce) instructions.
A/J mice were immunized initially intraperitoneally (IP) with the Aβ conjugate mixed with complete Freund's adjuvant. Fourteen days later, the mice were boosted IP with the Aβ conjugate mixed with phosphate buffered saline (PBS) at 14 day intervals. After six total boosts, the mice were finally boosted intravenously with Aβ conjugate mixed with incomplete Freund's adjuvant and fused 3 days later. Fusion of spleen cells with P3.653 myeloma cells was performed as described in Oi and Herzenberg, Selective Methods in Cellular Immunology, Mishell and Shigii, Eds., W. H. Freeman and Company, San Francisco, Chapter 17 (1980). Serum titers and initial screens were performed by the RIA method described below. Several clones were expanded to a 24-well plate and subjected to further analysis as described below. Clones of interest were produced in mouse ascites.
The RIA method used to screen serum bleeds and fusion hybridoma supernatants was based upon a method developed by Wang et al. (1977) J. Immunol. Methods 18:157-164. Briefly, the supernatant (or serum) was incubated overnight at room temperature on a rotator with 125I-labeled Aβ1-28 and Sepharose® 4B beads to which sheep anti-mouse IgG had been coupled via cyanogen bromide. The beads from each well were harvested onto glass fiber filter discs with a cell harvester and washed several times with PBS. The filter discs were then transferred to gamma tubes and the bound radioactivity was counted in a gamma counter.
All hybridomas were tested for binding to Aβ1-28 using the method described above in the initial screen, and then retested 3 days later. Aβ1-28 positive clones were further characterized for reactivity to 125I-labeled Aβ1-16 using the RIA method described above. No clones were found to bind Aβ1-16. In a peptide capture ELISA, all clones were found to react with Aβ13-28 while no clones reacted with Aβ17-28. Therefore, it was determined that all clones had an epitope within the junction region spanning amino acids 16 and 17.
Based on results of the above assays, several clones were expanded into 24 well plates. These clones were further characterized by saturation analysis. Supernatants at the 50% titer point (as determined by the RIA method described above) were added to wells containing Sepharose®-sheep anti-mouse IgG beads, a constant amount of 125I-labeled Aβ1-28, and varying amounts of unlabeled junction peptide or Aβ17-28. The concentration of cold peptide for 50% inhibition was determined for each antibody. For the Aβ17-28, no inhibition was seen at 100 ng/well for any clones. The 50% inhibition point for junction peptide ranged from 10-80 ng/well. The clones were also characterized based on reactivity in Western blots. Based on titer point, sensitivity (as determined by the 50% inhibition point), and reactivity on Western blot, several clones were produced in ascites. Antibodies from hybridoma designated 266 (the “266 antibody”) was selected for use as a capture antibody in the assays described below.
Supernatants from wells containing hybridoma cells were screened for antibody with the ability to capture 125I labeled Aβ 1-42 in solution by immunoprecipitation (Notebook 1101).
B. 2G3 Production
Antibody 2G3, specific for Aβ(x-40), was produced by injecting female A/J mice intraperitoneally with 100 μg immunogen per injection. The immunogen consisted of the peptide NH2-Cys-NH—CH2—(CH2)5—CO-GLMVGGVV-COOH (SEQ ID NO: 5), coupled to sheep anti-mouse IgG using maleimidohexanoyl-N-hydroxysuccinimide. The immunogen was emulsified with Freund's complete adjuvant for the first immunization, and all subsequent immunizations were with 100 μg of immunogen emulsified with Freund's incomplete adjuvant at approximately two week intervals.
Three days before fusion, a mouse was boosted with PBS solutions containing 50 μg immunogen intravenously and 50 μg intraperitoneally in PBS. The mouse was sacrificed, the spleen was removed, splenocytes were isolated and fused with the SP2/0 mouse myeloma using a modification of the method of Koehler and Milstein.
Supernatants from wells containing hybridoma cells were screened for the ability to produce antibody which recognizes Aβ(1-40) which had been coated onto an ELISA plate. “Positives” were further screened for their ability to capture 125I Aβ(1-40) in solution by immunoprecipitation.
C. Production of 21F12
Antibody 21F12, specific for Aβ(x≧41) was produced by immunizing A/J mice intraperitoneally with 100 μg of immunogen per injection. The immunogen consists of the synthetic peptide NH2-Cys-NH—CH2(CH2)5—CO-GLMVGGVVIA-COOH (SEQ ID NO: 4) coupled to sheep anti-mouse IgG using maleimidohexanoyl-N-hydroxysuccinimide (MHS). The immunogen was emulsified with Freund's complete adjuvant for the first immunization, and all subsequent immunizations were with 100 μg of immunogen emulsified with Freund's incomplete adjuvant at approximately two week intervals.
Three days before the fusion, a mouse was injected with 50 μg of immunogen each intravenously and intraperitoneally of immunogen in PBS. Three days post injection, the spleen was removed, splenocytes were isolated and fused with SP2/0 following a modification of the method of Koehler and Milstein.
D. Specificity of 2G3 and 21F12
The specificities of antibody 2G3 and 21F12 are demonstrated in FIGS. 1 and 2. In this assay, antibody 2G3 or 21F12 was coated into the wells of an ELISA plate by diluting the purified antibody to a concentration of 10 μg/ml in Well-coating Buffer (0.01 M PO4 pH 8.5), and pipetting 100 μl of the antibody solution into each well. The solution was left overnight at room temperature and then was removed by aspiration. The non-specific sites of the well were blocked by the addition of 200 μl 0.25% Human Serum albumin in PBS and incubated for at least one hour at room temperature. The blocking solution was removed and the wells were washed one time with wash buffer (Tris buffered Saline, 0.05% Tween 20).
Standards containing between 80-20,000 pg/ml of either Aβ(1-40) or Aβ(1-42) were then prepared by dilution in Specimen Diluent (1 mM PO4, 0.15 M NaCl, pH 7.4, 0.6% Bovine Serum albumin, globulin-free, 0.05% Triton X-405 and 0.05% Thimerosal), and 100 μl of each of these standards were added to the appropriate wells. The standards were incubated for one hour at room temperature, then aspirated and the wells washed four times with wash buffer.
100 μl of a second antibody (the reporter antibody) was added at a concentration of 0.5 μg/ml in specimen diluent. This reporter antibody is biotinylated 3D6 (which recognizes Aβ (1-5)) prepared by the reaction of antibody with NHS-biotin (Pierce). This was allowed to incubate one hour at room temperature, and then washed four times with wash buffer.
To each well, 100 μl of a 1/5000 dilution of avidin HRP (Vector Labs) was added and allowed to incubate one hour at room temperature. The wells were washed four times in wash buffer, and 100 μl of Slow TMB (Pierce) were added to each well and incubated fifteen minutes. The reaction was stopped by the addition of 25 μl of 2 M H2SO4 and the plates were read at 450-650 on a Vmax reader (Molecular Devices) (Notebook 1344).
As can be seen in FIG. 1, antibody 2G3 reacts strongly with Aβ(1-40), but has essentially no cross-reactivity with Aβ(1-42). In FIG. 2, it is shown that antibody 21F12 similarly has very high specificity, in this case for Aβ(1-42) over Aβ(1-40). At a concentration of 20,000 pg/ml less than 0.4% of cross reactivity is observed.
II. ELISA Assay
A. Binding of Capture Antibody to Microtiter Wells
A monoclonal antibody against Aβ(x≧41) or Aβ(x≦40) is diluted to a concentration of 10 μg/ml in a buffer containing 0.23 g/L NaH2PO4.H2O, 26.2 g/L Na2HPO4.7H2O, 1 g/L NaN3, pH 8.5. One hundred μl/well of this solution is then dispensed in a 96 well white Dynatech Microlite 2, 96 well flat-bottomed plate. The plates are sealed and incubated overnight at room temperature. Following coating, the remaining solution was aspirated and the non-specific binding sites are blocked with 200 μL per well of (NaH2PO4.H2O) 0.2 g/L, Na2HPO4.7H2O 0.8 g/L, human serum albumin (HSA) crystallized and lyophilized 2.5 g/L, pH 7.4. These plates are blocked by incubating for 1 hour at room temperature in the blocking solution.
B. Assay Protocol
The calibrators are prepared from a stock solution of Aβ1-42, 1 μg/ml, in DMSO. In specimen diluent ((NaH2PO4.H2O) 0.2 g/L, Na2HPO4.7H2O 2.16 g/L, NaN3 0.5 g/L, bovine serum albumin (BSA) (globulin free) 6 g/L, triton x-405 0.5 ml/L NaCl 8.5 g/L, pH 7.4.), the highest calibrator, 1000 pg/ml (10 μl Aβ1-42 stock (1 μg/ml DMSO) in 10 ml casein specimen diluent) is prepared. Sequential dilutions are made in specimen diluent to obtain 500, 250, 125, 62.5 and 31.25 pg/ml concentrations of Aβ1-42.
One hundred μL per well calibrators or samples are applied to the microtiter plates. The plates are sealed and incubated for 1 hour at room temperature. The plates are then washed three times with washing buffer (NaCl 80 g/L, KCl 3.85 g/L, Tris-HCl 31.75 g/L, tween-20 0.5 ml/L, pH 7.5).
Antibody is diluted in specimen diluent to 1 μg/ml and 100 μl is added per well. The plate is covered and incubated for 1 hour at room temperature. The plate is washed three times with washing buffer. The alkaline phosphatase affinity purified F(ab′)2 fragment donkey anti-rabbit IgG (H+L) (Jackson) is diluted 1:1000 in specimen diluent. One hundred μl/well is added. The plate is covered and incubated for 1 hour at room temperature. The plate is washed three times with washing buffer, then 100 μl/well of chemiluminescent substrate is added. The chemiluminescent substrate is prepared by diluting the chemiluminescent reagent, AMPPD (Tropix), and an enhancer, emerald green (Tropix), 1:1000 and 1:100 respectively in 1M diethanolemine buffer, pH 10, containing 1 mM MgCl2 and 0.2% NaN3. The plates are sealed and incubated for 10 to 15 minutes at room temperature. Solution is not aspirated. This time may have to be optimized for different antibody lots.
Chemiluminescence is read and expressed as relative chemiluminescence units (CLU) after 15 minutes using a Dynatech ML 1000.
III. Inhibition of APP Processing by the MDL 28170 Inhibitor
We first set out to reproduce the published results of Higaki et al. (1995) Neuron, 14:651-659 on the action of the compound MDL 28170 on APP processing, using human kidney 293 cells stably expressing APP695 with the Swedish FAD mutation (K695sw cells). Our experiments were done using a pulse-chase paradigm: K695sw cells were labeled for 2 hours with 35S-methionine and then chased for 2 hours in the presence of 200 μM MDL 28170. Aliquots of the chase media from treated and untreated cells were subjected to SDS-PAGE. No significant differences in the amounts of the major secreted cellular proteins were detected (FIG. 3, lanes 1, 2), suggesting that under the conditions of the experiment, MDL 28170 does not interfere with general protein secretion.
We next analyzed the chase media for changes in the amounts of α- and β-cleaved APPs, using antibodies specific for each form. Antibody 1736 specifically immunoprecipitates α-cleaved APPs (Haass et al. (1994) J. Biol. Chem. 269:17741-17748; Haass et al. (1995) Nature Med. 1:1291-1296). This antibody revealed an increase in α-APPs production upon treatment (FIG. 3, lanes 3, 4), indicating that MDL 28170 does not significantly inhibit α-secretase.
192sw specifically immunoprecipitates the β-cleaved APPs species ending with the Swedish mutant met596 (Knops et al. (1995) J. Biol. Chem. 270:2419-2422; Haass et al. (1995) Nature Med. 1:1291-1296). Immunoprecipitation with this antibody showed that MDL 28170 does not significantly inhibit β-secretase activity (FIG. 3, lanes 5, 6).
We next analyzed lysates of the K695sw cells for changes in cellular full-length APP and its C-terminal fragments using antibody C7, directed to the last 20 amino acids of APP (Podlisny et al. (1991) Am. J. Pathol. 138:1423-1435). This antibody precipitates N′- and N′/O′-glycosylated full-length APP and its 10 kDa C-terminal fragment (residues 613-695 of APP695) that remains membrane bound after α-secretase cleavage. Upon treatment with MDL 28170, a striking increase in the level of the 10 kDa C-terminal fragment was observed (FIG. 3, lanes 7, 8).
In K695sw cells, the 12 kDa C-terminal fragment (residues 597-695 of APP695) which remains membrane bound after β-secretase cleavage cannot be easily resolved and detected by antibody C7 (Citron et al., 1995). We therefore precipitated this fragment using antibody 1282 raised to synthetic Aβ(1-40). This antibody can precipitate the 12 kDa C-terminal but not the 10 kDa fragment, and therefore the faint 12 kDa band is not overshadowed by the much more abundant 10 kDa band. Whereas no 12 kDa fragment was detectable in untreated cells, this band was clearly observed upon treatment with MDL 28170 (FIG. 3, lanes 9, 10).
In summary, no significant inhibition of α- or β-secretase cleavage, but an increase in both the 10 and 12 kDa C-terminal fragments was observed upon treatment with MDL 28170, strongly supporting the role of this compound as a γ-secretase inhibitor which inhibits the conversion of the 10 and 12 kDa fragments to the p3 and Aβ peptides, respectively.
IV. MDL 28170 Inhibits the Production of Aβ(40) and p3(40) but not Aβ(42) and p3(42)
MDL 28170 had previously been shown to inhibit the secretion of Aβ and p3 and therefore had been suggested to inhibit γ-secretase. This inhibition was observed by immunoprecipitating media from treated cells with a polyclonal antibody raised to synthetic Aβ(1-40) (Higaki et al., (1995) Neuron, 14:651-659). Since the vast majority of secreted Aβ and p3 peptides end at amino acid 40, this experiment does not distinguish whether only the major γ-secretase cleavage at position 40 is inhibited.
To address this question, we performed pulse-chase experiments on K695sw cells using different doses of MDL 28170, followed by sequential immunoprecipitation of the same media first with 21F12, a monoclonal antibody raised against amino acids 33-42 of Aβ (described above) which specifically precipitates Aβ peptides ending at position 42, and then with antibody 1282 which precipitates all forms of Aβ and p3 (Haass et al., (1992b) Nature 359:322-325) (FIG. 4A). Interestingly, 21F12 precipitated not only Aβ, but also p3 peptides, thus demonstrating the existence of secreted p3(42), which had not been described before. Total Aβ and total p3 were strongly and significantly decreased with doses of MDL 28170>50 μM (e.g., at 200 μM p<0.001). In contrast, Aβ(42) and p3(42) showed a bell-shaped dose response curve, with only a small and insignificant decrease at 200 μM, the dose used by Higaki et al. (1995) Neuron, 14:651-65.9, and the highest dose tested here. Using MDL 28170 at 200 μM, the experiment was repeated four times and the results were quantitated by phosphorimaging (FIG. 4B). These data indicate that under the conditions described above the differential effect is significant for both Aβ and p3.
V. Differential Inhibition is Accomplished Under a Number of Conditions
To make sure that the differential effect observed in Aβ(40)/Aβ(42) and p3(40)/p3(42) precipitations is meaningful, we performed a number of control experiments using the K695sw cells. First we treated K695sw cells in a 2 h pulse 2 h chase paradigm with 1 μM of the phorbolester PDBu, which has been shown to decrease total Aβ but increase total p3, probably by diverting βAPP substrate from the β-secretase pathway to the α-secretase route (Buxbaum et al., (1993) “Protein phosphorylation inhibits production of alzheimer amyloid β/A4 peptide,” Proc. Natl. Acad. Sci. USA 90:9195-9198; Hung et al., (1993) “Activation of protein kinase C inhibits cellular production of the amyloid β-protein,” J. Biol. Chem. 268:22959-22962). This effect should be independent of the subsequent γ-secretase cleavage and thus the 40 and 42 forms of each metabolite should be equally decreased or increased if the immunoprecipitation paradigm used here works correctly. Indeed, when conditioned media of PDBu-treated cells was precipitated with 21F12, the expected decrease in Aβ(42) and increase in p3(42) were observed indicating that Aβ(42) immunoprecipitation signal does reflect changes in the amounts of precipitable material. Subsequent immunoprecipitation with R1282 shows the same effect for total Aβ and total p3 (FIG. 5A).
The statement that Aβ(42) and p3(42) are not decreased by MDL 28170 depends critically on the quality of the 21F12 antibody. To confirm the effects observed with this antibody, two other previously published Aβ(42)-specific antibodies were used in the pulse chase paradigm with MDL 28170 at 200 μM. The monoclonal antibody BC05 has been extensively used in ELISA assays to detect Aβ(42) (Asami-Odaka et al., (1995) Biochemistry 34:10272-10278; Gravina et al., (1995) J. Biol. Chem. 270:7013-7016; Suzuki et al., (1994) Science 264:1336-1340).
When media from MDL 28170-treated K695sw cells was precipitated with this antibody, we observed an actual increase in both Aβ(42) and p3(42). The subsequent precipitation with R1282 showed the usual decrease in total Aβ and p3 (FIG. 5B).
The polyclonal antibody C42 has also been shown to be specific for Aβ(42) (Saido et al., (1994) Spatial resolution of the primary β-amyloidogenic process induced in postischemic hippocampus. J. Biol. Chem. 269:15253-15257). Likewise, this antibody did not show a decrease in Aβ(42) and p3(42) upon treatment whereas the subsequent precipitation with R1282 showed the usual decrease in total Aβ and p3 (FIG. 5C).
The decrease in Aβ(40) and p3(40) was also found when the monoclonal antibody, 2G3 (described above) specific for the free carboxyl-terminus of Aβ(40) and p3(40) was used to precipitate first, followed by 21F12 (FIG. 5D). As expected, the differential inhibition of Aβ production by MDL 28170 was also detected when the precipitations were carried out not sequentially (as described above) but in parallel after the standard pulse-chase. That is, aliquots of media from treated cells and from untreated cells were precipitated with 21F12 for the Aβ42 forms and other aliquots were precipitated with antibody 1282 for total Aβ and total p3. This parallel precipitation produced the same result as the sequential precipitations described above (FIG. 5E).
In summary, three different Aβ(42) end-specific antibodies show that MDL 28170 does not strongly decrease Aβ(42) and p3(42) production whereas a monoclonal antibody to Aβ(40) and p3(40) and different polyclonal antibodies to total Aβ and p3 show a strong decrease. Finally, immunoprecipitation with 21F12 followed by antibody 1282 again revealed this differential inhibition when the inhibitor (100 μM) was applied during a 3 h labeling period instead of in a pulse-chase format (FIG. 5F).
VI. Differential Inhibition is Observed in Several Cell Lines
To check whether the differential inhibition is specific for K695sw, three additional cell lines were treated in the standard 2 h pulse-2 h chase paradigm, and the conditioned media were precipitated first with 21F12 and then with antibody 1282. The kidney cell line K695717I expresses APP695 carrying the 717I mutation. This line was chosen because it produces increased levels of Aβ(42) due to the mutation (Suzuki et al., (1994) Science 264:1336-1340). At 200 μM MDL 28170, no decrease of Aβ(42) and p3(42) was observed, whereas Aβ(40) and p3(40) were substantially reduced. (FIG. 6A). The Chinese Hamster ovary cell line CHO695 transfected with wild-type βAPP695 cDNA was treated with 200 μM MDL 28170 and only a slight decrease of Aβ(42) and p3(42) was observed, whereas Aβ(40) and p3(40) were substantially reduced. (FIG. 6B). The human neuroblastoma cell-line SKN695 expressing wild type βAPP695 was treated with 200 μM MDL 28170 (FIG. 6C). While Aβ(42) and p3(42) were slightly increased, total Aβ and total p3 were strongly decreased. Thus, differential inhibition of Aβ(42) vs Aβ(40) and p3(42) vs p3(40) production is not only observed in K695sw but also in a cell line with an Alzheimer's disease linked βAPP717 missense mutation, in a hamster cell line and in a human neural cell line expressing wild type βAPP.
VII. Experimental Procedures
A. Cell Lines
All transfected cell lines described here carry derivatives of pCMV695, a plasmid carrying APP695 under control of the CMV promoter (Selkoe, (1988) Proc. Natl. Acad. Sci. 85:7341-7345) K695sw are human embryonic kidney 293 cells stably transfected with a construct carrying the AD-linked double (“Swedish”) mutation K595N/M596L (Citron et al. (1992) Nature, 360:672-674); K695717I are 293 cells stably transfected with APP695 carrying the mutation V717I (valine to isoleucine at position 717 in the APP770 numbering system). CHO695 are Chinese hamster ovary cells (CHO) stably transfected with pCMV695 (Oltersdorf (1990) J. Biol. Chem. 265:4432-4437) SKN695 are SK-N-SH human neuroblastoma cells stably transfected with pCMV695.
B. Pulse-Chase Experiments and Immunoprecipitations
To analyze the effect of MDL 28170 on the processing of APP, cells were grown to confluence in two 10 cm dishes, pulse-labeled with 600 μCi of [35S]-methionine in 4 ml of serum-free medium for 2 hours and then chased for 2 hours with 4 ml medium containing 10% fetal bovine serum and the indicated final concentration of MDL 28170 (initially dissolved at 200 mM. in DMSO). Control dishes were treated with DMSO alone.
Conditioned media and cell lysates were analyzed by immunoprecipitation, as described (Haass, et al. (1992) Nature 359:322-325). Polyclonal antibody R1736 to residues 595-611 of APP695 was used to precipitate α-APPs (Haass, (1994) J. Biol. Chem. 269:17741-17748). This antibody recognizes an epitope that is specific for the free COOH-terminus of α-cleaved APPs. Polyclonal antibody R1282 was generated to synthetic Aβ1-40 (Haass, et al. (1992) Nature 359:322-325). This antibody precipitates total Aβ and p3 (and small, variable amounts of APPs) from the media of cultured cells (Haass, et al. (1992) Nature 359:322-325). The monoclonal antibody 2G3 was raised to peptide C(Aminoheptanoic acid)GLMVGGVV [(SEQ ID NO: 5) and specifically precipitates Aβ(40) and p3(40). Twenty μg of this antibody were used to immunoprecipitate the chase media of 2 dishes. The monoclonal antibody 21F12 was raised to peptide C(Aminoheptanoic acid)GLMVGGVVIA (SEQ ID NO: 4) and specifically precipitates Aβ(42) and p3(42). Twenty μg of this antibody were used to immunoprecipitate the chase media of 2 dishes. The monoclonal antibody BC05 specifically detects Aβ(42) and p3(42) (Suzuki et al., (1994) Science 264:1336-1340). The polyclonal antibody C7 against the last 20 residues of the APP cytoplasmic tail (Podlisny, (1991) Am. J. Pathol. 138:1423-1435) precipitates N′- and N⇑ plus O⇑-glycosylated full-length APP as well as its C-terminal proteolytic fragments. The antibody sw192 (Knops et al. (1995) J. Biol. Chem. 270:2419-2422; Haass et al. (1995) Nature Med.1:1291-1296) specifically precipitates β-cleaved APPs carrying the Swedish mutation.
SDS-PAGE of immunoprecipitates of cell extracts or of Aβ from media was carried out on 10-20% Tris-Tricine gels (Novex), whereas APPs precipitates were electrophoresed on 10% SDS-polyacrylamide Tris Glycine gels. All quantitations were performed with a Phosphorimager 400A using Image-QuaNT software (Molecular Dynamics). It should be noted the pulse/chase-immunoprecipitation method allows one to assess any changes in Aβ and p3 simultaneously in the same assay, with each of these peptides being visualized directly in the electrophoretic gel.
VIII. Human Neurons
Human neurons were cultured as previously described (Seubert et al. Nature (1992) 359:325-327) except that the cells were seeded into 6-well plates in neuronal medium without fetal bovine serum but supplemented with B27 (Gibco). Cells were cultured for 2-3 weeks in serum free medium prior to use.
PDAPP mouse brain cells from 16 day old fetal cerebral cortex were cultured following the protocol for human neurons except the cells were seeded into 24-well plate clusters in neuronal medium with 50 fetal bovine serum (Sigma) and 5% Chang's supplement (Irvine Scientific). Cells were cultured for 5-7 days prior to being used in experiments.
The procedure for examining the effects of substances on Aβ production is as follows. Fresh medium is added to the culture wells and then collected after ˜24 (8-30) hrs. This is the “control” sample from each well that the treated sample will be compared to. Fresh medium, containing the substance to be tested is then added and again harvested after a further ˜24 hr (8-30) incubation. After collection of this “treated” sample, a cytotoxicity assay is performed on the cells. To perform the cytotoxicity assay, cells are incubated in media containing thiazolyl blue (MTT, Sigma) at 1 mg/ml for 15 minutes. The media are then discarded, and the precipitates are analyzed by solubilization in a buffer containing 50% DMF and 20% SDS. The solubilized dye was quantitated on a Molecular Devices Vmax.
Control and treated samples of culture media are assayed for total Aβ using a sandwich ELISA consisting of two monoclonal antibodies. The first antibody 266, specific to amino acids 13-28 of Aβ, is used as a capture antibody (Seubert et al., Nature, supra). The second antibody, 3D6 which is specific to amino acids 1-5 of Aβ, was biotinylated and served as a reporter antibody. The 3D6 biotinylation procedure employed the manufacturer's protocol for NHS-biotin (Pierce) labeling of immunoglobulins, except 100 mM sodium bicarbonate, pH 8.5, buffer was used. The 3D6 antibody does not recognize secreted APP or full-length APP but does recognize Aβ species that begin at position 1.
The samples were also assayed for Aβ(42) with an Aβ(42) specific sandwich ELISA that employed the monoclonal antibody 21F12, which was generated against amino acids 33-42 of Aβ, as the capture antibody. This antibody is specific for longer forms of Aβ since it does not cross-react with Aβ(1-40) in ELISA or competitive radioimmunoassay (RIA). Biotinylated 3D6 is also the reporter antibody in this assay.
The 266 and 21F12 mAbs were coated at 10 μg/ml into 96-well immunoassay plates (Costar) overnight at room temperature. The plates were aspirated and blocked with 0.25% human serum albumin PBS buffer for at least 1 hour at room temperature, then stored desiccated at 4° C. until use. The samples and standards were added to the plates and incubated at room temperature for 1.5 hours.
The biotinylated 3D6 was diluted to 0.5 μg/ml, and incubated in the wells for 1 hour at room temperature. The plates were washed 3 times with wash buffer (Tris buffered saline, 0.05% Tween 20) between each step of the assay. Streptavidin-alkaline phosphatase (Boehringer Mannheim), diluted 1:1000, was added to the wells for the total Aβ assay, and avidin-HRP (Vector) diluted 1:4000 was added to the wells for the Aβ(42) assay. These conjugates were incubated for 1 hour at room temperature. For the total Aβ assay the fluorometric substrate 4-methyl-umbelliferyl phosphate was added to the wells for 30 minutes, then read in a Millipore Cytofluor 2350 fluorometer. The calorimetric substrate, Slow TMB-ELISA (Pierce), was added for the Aβ(42) assay and allowed to react for 15 minutes, after which the enzymatic reaction was stopped with 2N H2SO4. The plates were read on a Molecular Devices Vmax.
Percent inhibition for both total Aβ and Aβ(42) is defined as:
(1−((treated/controlt)/(untreated/controlu)))×100%,
where treated=value from treated cells
    • controlt=value from the same treated well for the 24 hr period prior to testing
    • untreated=value from well which received no test substance
    • controlu=value from the same untreated well for the 24 hr prior to testing
It is optimal to divide the values of the treated and untreated samples by their respective values for the 24 hrs prior to the 24 hr test period, as the act of changing the media alone can effect a 5-10% reduction in Aβ production (comparing the untreated to controlu).
Several compounds were screened for their ability to inhibit both total Aβ and Aβ(42). Results are shown in FIGS. 13A-13O.
IX. PDAPP Construct
A cDNA/genomic APP construct containing introns 6, 7 and 8 is prepared by combining APP cDNA encoding exons 1-6 and 9-18 with genomic APP sequences encoding introns 6, 7 and 8, and exons 7 and 8 (see FIGS. 8A-8B). In order to create a splicing cassette small enough for convenient insertion in a pUC vector, two deletions in intronic sequences are made. A deletion is made in intron 6 from position 143 of intron 6 to the BamHI site located upstream of the beginning of exon 7 (1658 bp before the beginning of exon 7). Another deletion is made in intron 8 from the first BamHI site in intron 8 to a site at 263 bp before the beginning of exon 9. To avoid confusion, these truncated forms of APP introns 6 and 8 are referred to herein as intron Δ6 and Δ8. BamHI sites are engineered at the sites of these deletions, so that they are marked by the presence of BamHI sites. In this construct, referred to as PDAPP, exons 7 and 8 and intron 7 are intact genomic sequences, except that the unique XhoI site in intron 7 is destroyed.
DNA fragments containing the truncated introns are generated as follows: a BamHI site is engineered 143 bp into intron 6 nucleotide by PCR-mutagenesis (“Mutagenesis by PCR” in PCR Technology: Current Innovations (Griffith and Griffith, eds., CRC Press, 1994) pages 69-83) and another BamHI site is engineered by PCR mutagenesis 263 bp prior to the beginning of exon 9. These sites are engineered into separate APP genomic DNA clones containing the junctions of exon 6 and intron 6, and intron 8 and exon 9, respectively, resulting in modified APP genomic DNA clones.
The entire cassette is assembled in the APP cDNA clone as follows (FIG. 9). The 889 bp BamHI to XcmI fragment of APP cDNA containing exons 1 through 5 and part of exon 6 (including nucleotides 1 to 843 of FIG. 10 (SEQ ID NO: 2)) is cloned into a vector containing BamHI and XhoI sites downstream from the insertion site to make APP770x-oligo-x. APP770x-oligo-x is then cut with XcmI and BamHI. Then two fragments are obtained from the modified APP genomic DNA clone containing the junction of exon 6 and intron 6 described above by cutting with XcmI and BamHI. The resulting 34 bp fragment from the XcmI in exon 6 to the XcmI in intron 6, and 131 bp fragment from the XcmI in intron 6 to the artificially created BamHI site at position 143 bp of intron 6 are ligated into APP770x-oligo-x in a three-way ligation step to make APP 770x-E6oligo-x. The orientation of the fragments are confirmed by sequencing. APP770x-E6oligo-x is then cut with BamHI and XhoI. Then the 313 bp BamHI and XhoI fragment from the modified APP genomic DNA clone containing the junction of intron 8 and exon 9 is ligated into APP770x-E6oligo-x to make APP770xE6E9x.
APP770xE6E9x is then cut with BamHI and the 6.8 kb BamHI fragment of APP genomic DNA encoding the KPI and OX-2 domains (exons 7 and 8) is inserted at this site. This fragment starts at the BamHI site 1658 bp upstream of the start of exon 7 and extends to the first BamHI site in intron 8. This BamHI fragment is obtained from a lambda phage genomic clone encoding this portion of the APP gene, that was obtained from a Human Placental genomic library in the Lambda FIXII vector obtained from Stratagene. This BamHI fragment originally contained an XhoI site which was destroyed by cutting fill in and relegation. The locations of the deletions are diagramed in FIG. 11. This clone, containing exons 1-8 and part of 9, and introns 6, 7 and 8, is termed the “APP splicing cassette.” The APP splicing cassette is cut out with NruI and XhoI and used to replace the NruI to XhoI cDNA fragment of APP cDNA bearing a Hardy mutation. This mutant form of APP cDNA is produced by converting the G at nucleotide position 2145 to T by site directed mutagenesis. This changes the encoded amino acid from Val to Phe. The resulting construct is a combination cDNA/genomic APP “minigene.”
Sequencing of the 6.8 kb BamHI fragment containing APP exons 7 and 8 derived from the APP genomic clone used to generate this construct showed that intron 7 is 2.6 kb long, and that the first BamHI site in intron 8, the upstream site of the deletion in intron 8 engineered into the APP minigene construct, is 2329 bp downstream from the end of exon 8. This does not coincide with the restriction map of the APP gene published by Yoshikai et al. (1990) and Yoshikai et al. (1991). Comparison of their map to our sequence indicates that Yoshikai et al. switched the order of two EcoRI fragments in their restriction mapping. The 1.60 kb EcoRI fragment containing exon 8 is actually upstream of the 1.48 kb EcoRI fragment and the 1.48 kb EcoRI fragment Yoshikai et al. mapped into intron 7 is actually in intron 8. We have confirmed this location for the EcoRI fragment containing exon 8 by sizing of PCR generated fragments from human DNA.
This APP minigene is operatively linked to the PDGF-β promoter to provide expression of the APP cDNA/genomic construct in mammalian cells. The PDGF β-chain 5′ flanking sequence is inserted upstream of the NruI site at the beginning of the APP minigene. This fragment includes 1.3 kb upstream of the transcription initiation site, where the PDGF-β promoter resides, and approximately 70 bp of 5′ untranslated region, ending at the AurII site (Higgins et al. (1994)). The late SV40 polyadenylation signal, carried on a 240 bp BamHI to BclI fragment, is added downstream of the APP minigene. This construct, combining the PDGF-β promoter, the APP splicing cassette, a Hardy mutation, and the SV40 polyadenylation signal is referred to as PDAPP (FIG. 12).
The present invention provides a novel screening method for determining whether a compound alters the production of Aβ(x-≧41) and/or Aβ(x-≦40) or total Aβ. While specific examples have been provided, the above description is illustrative and not restrictive. Many variations of the invention will become apparent to those of skill in the art upon review of this specification. The scope of the invention should, therefore, be determined not with reference to the above description, but instead should be determined with reference to the appended claims along with their full scope of equivalents.
All publications and patent documents cited in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication or patent document were so individually denoted.

Claims (5)

1. A method for determining a compound is a compound that inhibits production of Aβ(42) and Aβ(40) separately:
obtaining control values which are characteristic of the level of Aβ42 peptide and total Aβ or Aβ40 peptide in a sample from a non-human animal used as a model of Alzheimer's disease;
administering the compound to the non-human animal used as a model of Alzheimer's disease;
measuring an amount of Aβ42 peptide in a sample from the non-human animal administered the compound; and
measuring an amount of total Aβ or Aβ40 peptide in a sample from the non-human animal administered the compound;
wherein the measured amounts in comparison with the control values indicate differential inhibition of Aβ (42) and Aβ (40)thus indicating the compound is a compound that inhibits production of Aβ(42) and Aβ(40) separately.
2. The method of claim 1 wherein the non-human animal is a rodent.
3. The method of claim 2 wherein the rodent is a mouse.
4. The method of claim 1 wherein the non-human animal is a rodent and expresses the Swedish mutation of human beta-amyloid precursor protein (APP).
5. A method for determining a compound is a compound that inhibits production of Aβ (40) and not Aβ (42):
obtaining control values which are characteristic of the level of Aβ42 peptide and total Aβ or Aβ40 peptide in a sample from a non-human animal used as a model of Alzheimer's disease;
administering the compound to the non-human animal used as a model of Alzheimer's disease;
measuring an amount of Aβ42 peptide in a sample from the non-human animal administered the compound;
measuring an amount of total Aβ or Aβ40 peptide in a sample from the non-human animal administered the compound; and
wherein the control values in comparison with the measured amounts indicate the compound is a compound that inhibits production of Aβ40 and not Aβ42.
US10/335,035 1992-07-10 2002-12-30 Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal Expired - Fee Related US7993627B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/335,035 US7993627B2 (en) 1992-07-10 2002-12-30 Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US91164792A 1992-07-10 1992-07-10
US96597292A 1992-10-26 1992-10-26
US08/079,511 US5766846A (en) 1992-07-10 1993-06-17 Methods of screening for compounds which inhibit soluble β-amyloid peptide production
US08/665,649 US6610493B1 (en) 1993-06-17 1996-06-18 Screening compounds for the ability to alter the production of amyloid-β peptide
US10/335,035 US7993627B2 (en) 1992-07-10 2002-12-30 Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US08/665,649 Continuation US6610493B1 (en) 1992-07-10 1996-06-18 Screening compounds for the ability to alter the production of amyloid-β peptide
US08/665,649 Division US6610493B1 (en) 1992-07-10 1996-06-18 Screening compounds for the ability to alter the production of amyloid-β peptide

Publications (2)

Publication Number Publication Date
US20030148392A1 US20030148392A1 (en) 2003-08-07
US7993627B2 true US7993627B2 (en) 2011-08-09

Family

ID=24670990

Family Applications (2)

Application Number Title Priority Date Filing Date
US08/665,649 Expired - Fee Related US6610493B1 (en) 1992-07-10 1996-06-18 Screening compounds for the ability to alter the production of amyloid-β peptide
US10/335,035 Expired - Fee Related US7993627B2 (en) 1992-07-10 2002-12-30 Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US08/665,649 Expired - Fee Related US6610493B1 (en) 1992-07-10 1996-06-18 Screening compounds for the ability to alter the production of amyloid-β peptide

Country Status (8)

Country Link
US (2) US6610493B1 (en)
EP (1) EP0906577B2 (en)
JP (1) JP3694033B2 (en)
AT (1) ATE407362T1 (en)
AU (1) AU3572797A (en)
CA (1) CA2258348C (en)
DE (1) DE69738961D1 (en)
WO (1) WO1997048983A1 (en)

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6610493B1 (en) 1993-06-17 2003-08-26 Brigham And Women's Hospital Screening compounds for the ability to alter the production of amyloid-β peptide
US7427392B1 (en) * 1994-11-14 2008-09-23 Elan Pharmaceuticals, Inc. Methods for aiding in the diagnosis of alzheimer's disease by measuring amyloid-β peptide (x-≧41) and tau
GB9701684D0 (en) 1997-01-28 1997-03-19 Smithkline Beecham Plc Novel compounds
US8173127B2 (en) * 1997-04-09 2012-05-08 Intellect Neurosciences, Inc. Specific antibodies to amyloid beta peptide, pharmaceutical compositions and methods of use thereof
US7964192B1 (en) 1997-12-02 2011-06-21 Janssen Alzheimer Immunotherapy Prevention and treatment of amyloidgenic disease
US7179892B2 (en) * 2000-12-06 2007-02-20 Neuralab Limited Humanized antibodies that recognize beta amyloid peptide
US7790856B2 (en) 1998-04-07 2010-09-07 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize beta amyloid peptide
US20080050367A1 (en) 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
TWI239847B (en) 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US6787523B1 (en) * 1997-12-02 2004-09-07 Neuralab Limited Prevention and treatment of amyloidogenic disease
US7700751B2 (en) 2000-12-06 2010-04-20 Janssen Alzheimer Immunotherapy Humanized antibodies that recognize β-amyloid peptide
CA2500462A1 (en) * 2001-12-26 2004-03-25 Universidad De Zaragoza Polyclonal antibodies, preparation method thereof and use of same
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
ES2201929B1 (en) * 2002-09-12 2005-05-16 Araclon Biotech, S.L. POLYCLONAL ANTIBODIES, METHOD OF PREPARATION AND USE OF THE SAME.
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
WO2004069182A2 (en) * 2003-02-01 2004-08-19 Neuralab Limited Active immunization to generate antibodies to soluble a-beta
TWI374893B (en) * 2003-05-30 2012-10-21 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
WO2005018424A2 (en) * 2003-08-18 2005-03-03 Research Foundation For Mental Hygiene, Inc. Antibodies specific for fibrillar amyloid and a procedure to detect fibrillar amyloid deposits
KR20070073778A (en) * 2004-10-28 2007-07-10 산코 준야쿠 가부시키가이샤 Method of examining alzheimer's disease and diagnostic reagent
WO2006053251A2 (en) * 2004-11-12 2006-05-18 Pfizer, Inc. Method of measuring amyloid-beta peptides
JP2008523815A (en) 2004-12-15 2008-07-10 エラン ファーマ インターナショナル リミテッド Humanized amyloid beta antibody for use in improving cognition
US20060241038A1 (en) * 2005-04-20 2006-10-26 Eisai Co., Ltd. Therapeutic agent for Abeta related disorders
BRPI0619249A2 (en) 2005-11-30 2011-09-20 Abbott Lab anti-globulin-ß antibodies, antigen-binding fractions thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of making said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
KR101667623B1 (en) 2005-11-30 2016-10-19 애브비 인코포레이티드 Monoclonal antibodies against amyloid beta protein and uses thereof
EP2361638B1 (en) 2005-12-12 2014-01-15 AC Immune S.A. A beta 1-42 specific monoclonal antibodies with therapeutic properties
US8784810B2 (en) 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
CA2657681C (en) 2006-07-14 2019-03-19 Ac Immune S.A. Humanized antibodies against beta amyloid protein
US8455626B2 (en) * 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
EP2124952A2 (en) 2007-02-27 2009-12-02 Abbott GmbH & Co. KG Method for the treatment of amyloidoses
US8003097B2 (en) 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
US8613923B2 (en) 2007-06-12 2013-12-24 Ac Immune S.A. Monoclonal antibody
US8048420B2 (en) 2007-06-12 2011-11-01 Ac Immune S.A. Monoclonal antibody
DK2182983T3 (en) 2007-07-27 2014-07-14 Janssen Alzheimer Immunotherap TREATMENT OF AMYLOIDOGENIC DISEASES WITH HUMANIZED ANTI-ABETA ANTIBODIES
PL2238166T3 (en) 2007-10-05 2014-07-31 Genentech Inc Use of anti-amyloid beta antibody in ocular diseases
US20100297012A1 (en) * 2007-10-05 2010-11-25 Andrea Pfeifer Humanized antibody
JO3076B1 (en) 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap Immunotherapy regimes dependent on apoe status
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
EP2258398A1 (en) 2009-05-26 2010-12-08 Araclón Biotech, S. L. Albumin-amyloid peptide conjugates and uses thereof
US9192585B2 (en) 2009-07-31 2015-11-24 Cognition Therapeutics, Inc. Inhibitors of cognitive decline
WO2011106785A2 (en) * 2010-02-26 2011-09-01 Cognition Therapeutics, Inc. Methods of identifying agents effective to treat cognitive decline and diseases associated therewith
CN102933601B (en) 2010-04-15 2016-06-08 Abbvie公司 Amyloid beta is in conjunction with albumen
RU2607368C2 (en) 2010-07-30 2017-01-10 Ац Иммуне С.А. Safe and functional humanized antibodies
US9062101B2 (en) 2010-08-14 2015-06-23 AbbVie Deutschland GmbH & Co. KG Amyloid-beta binding proteins
MX368755B (en) 2014-01-31 2019-10-15 Cognition Therapeutics Inc Isoindoline compositions and methods for treating neurodegenerative disease.
CA3061787A1 (en) 2017-05-15 2018-11-22 Cognition Therapeutics, Inc. Compositions for treating neurodegenerative diseases

Citations (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4271140A (en) 1978-01-23 1981-06-02 Baxter Travenol Laboratories, Inc. Method and composition for double receptor, specific binding assays
EP0123527A2 (en) 1983-04-19 1984-10-31 The Wistar Institute HLA D typing assay
US4486530A (en) 1980-08-04 1984-12-04 Hybritech Incorporated Immunometric assays using monoclonal antibodies
EP0171496A2 (en) 1984-08-15 1986-02-19 Research Development Corporation of Japan Process for the production of a chimera monoclonal antibody
EP0173494A2 (en) 1984-08-27 1986-03-05 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by DNA splicing and expression
WO1986001533A1 (en) 1984-09-03 1986-03-13 Celltech Limited Production of chimeric antibodies
EP0184187A2 (en) 1984-12-04 1986-06-11 Teijin Limited Mouse-human chimaeric immunoglobulin heavy chain, and chimaeric DNA encoding it
WO1987002671A1 (en) 1985-11-01 1987-05-07 International Genetic Engineering, Inc. Modular assembly of antibody genes, antibodies prepared thereby and use
JPS62100291A (en) 1985-10-28 1987-05-09 Teijin Ltd Gene fragment and plasmid
US4666829A (en) 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
EP0391714A2 (en) 1989-04-05 1990-10-10 BRIGHAM &amp; WOMEN'S HOSPITAL Diagnostic method for Alzheimer's disease: examination of non-neural tissue
WO1990012870A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Monoclonal antibody to amyloid peptide
WO1990012871A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Cerebrovascular amyloid protein-specific monoclonal antibody sv17-6e10
WO1991004339A1 (en) 1989-09-18 1991-04-04 California Biotechnology Inc. Assays and reagents for amyloid deposition
EP0444856A2 (en) 1990-02-26 1991-09-04 Albert Einstein College Of Medicine Of Yeshiva University Diagnostic assay for Alzheimer's disease
WO1991016628A1 (en) 1990-04-24 1991-10-31 The Regents Of The University Of California Purification, detection and methods of use of protease nexin-2
WO1991019810A1 (en) 1990-06-15 1991-12-26 California Biotechnology Inc. Transgenic non-human mammal displaying the amyloid-forming pathology of alzheimer's disease
WO1992000521A1 (en) 1990-06-29 1992-01-09 Case Western Reserve University Diagnostic and prognostic methods based on soluble derivatives of the beta amyloid protein precursor
WO1992009699A1 (en) 1990-11-27 1992-06-11 L.F. Will & Cie (Belgique) Monoclonal antibody for use in diagnosing alzheimer's disease, hybridoma secreting said antibody and preparation method therefor
US5134062A (en) 1988-03-22 1992-07-28 Cornell Research Foundation, Inc. Diagnosis of neuronal disorders and screening potential therapeutic agents therefor
WO1992013069A1 (en) 1991-01-21 1992-08-06 Imperial College Of Science, Technology & Medicine Test and model for alzheimer's disease
WO1993001817A1 (en) 1991-07-25 1993-02-04 Charles Lwanga Ssali Antiviral compositions comprising fusidic acid, l-ascorbic acid and salicylic acid and derivatives
US5200339A (en) 1990-08-17 1993-04-06 Abraham Carmela R Proteases causing abnormal degradation of amyloid β-protein precursor
WO1993008264A1 (en) 1991-10-17 1993-04-29 Valio Bioproducts Ltd. Colostrum fraction, preparation and use thereof as cell culture media supplement
US5221607A (en) 1989-09-18 1993-06-22 Scios Nova Inc. Assays and reagents for amyloid deposition
WO1993014200A1 (en) 1992-01-07 1993-07-22 Tsi Corporation Transgenic animal models for alzheimer's disease
US5234814A (en) 1989-06-01 1993-08-10 Du Pont Merck Pharmaceutical Company Diagnostic assay for alzheimer's disease
WO1993016101A1 (en) 1992-02-05 1993-08-19 Athena Neurosciences, Inc. Beta-amyloid peptide production inhibitors and methods for their identification
WO1993021526A1 (en) 1992-04-15 1993-10-28 Athena Neurosciences, Inc. METHODS AND COMPOSITIONS FOR MONITORING CELLULAR PROCESSING OF β-AMYLOID PRECURSOR PROTEIN
WO1994000569A1 (en) 1992-06-18 1994-01-06 Genpharm International, Inc. Methods for producing transgenic non-human animals harboring a yeast artificial chromosome
WO1994001772A1 (en) 1992-07-13 1994-01-20 The Children's Medical Center Corporation SCREEN FOR ALZHEIMER'S DISEASE THERAPEUTICS BASED ON β-AMYLOID PRODUCTION
WO1994010569A1 (en) 1992-10-26 1994-05-11 Schenk Dale B METHODS AND COMPOSITIONS FOR THE DETECTION OF SOLUBLE β-AMYLOID PEPTIDE
WO1994023049A2 (en) 1993-04-02 1994-10-13 The Johns Hopkins University The introduction and expression of large genomic sequences in transgenic animals
US5455169A (en) 1992-06-04 1995-10-03 Alzheimer's Institute Of America, Inc. Nucleic acids for diagnosing and modeling Alzheimer's disease
WO1996015452A1 (en) 1994-11-14 1996-05-23 Athena Neurosciences, Inc. METHODS FOR AIDING IN THE DIAGNOSIS OF ALZHEIMER'S DISEASE BY MEASURING AMYLOID-β PEPTIDE (X- ≥41) AND TAU
US5547841A (en) 1987-10-08 1996-08-20 The Mclean Hospital Corporation In vitro method for screening for drugs that inhibit production or degradation of human A4-amyloid
US5593846A (en) 1992-07-10 1997-01-14 Athena Neurosciences Methods for the detection of soluble β-amyloid peptide
US5604102A (en) 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
US5605811A (en) 1992-10-26 1997-02-25 Athena Neurosciences, Inc. Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
WO1997010601A1 (en) 1995-09-14 1997-03-20 Samsung Electronics Co., Ltd. Refresh strategy for drams
WO1997048983A1 (en) 1996-06-18 1997-12-24 Athena Neurosciences, Inc. SCREENING COMPOUNDS FOR THE ABILITY TO ALTER THE PRODUCTION OF AMYLOID-β PEPTIDE (x-≥41)
US5750349A (en) 1993-01-25 1998-05-12 Takeda Chemical Industries Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5837672A (en) 1992-07-10 1998-11-17 Athena Neurosciences, Inc. Methods and compositions for the detection of soluble β-amyloid peptide
US5850003A (en) 1993-10-27 1998-12-15 Athena Neurosciences Transgenic rodents harboring APP allele having swedish mutation
US5955317A (en) 1993-01-25 1999-09-21 Takeda Chemical Industries, Ltd. Antibodies to β-amyloids or their derivatives and use thereof

Patent Citations (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4271140A (en) 1978-01-23 1981-06-02 Baxter Travenol Laboratories, Inc. Method and composition for double receptor, specific binding assays
US4486530A (en) 1980-08-04 1984-12-04 Hybritech Incorporated Immunometric assays using monoclonal antibodies
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
EP0123527A2 (en) 1983-04-19 1984-10-31 The Wistar Institute HLA D typing assay
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US4736866B1 (en) 1984-06-22 1988-04-12 Transgenic non-human mammals
EP0171496A2 (en) 1984-08-15 1986-02-19 Research Development Corporation of Japan Process for the production of a chimera monoclonal antibody
EP0173494A2 (en) 1984-08-27 1986-03-05 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by DNA splicing and expression
WO1986001533A1 (en) 1984-09-03 1986-03-13 Celltech Limited Production of chimeric antibodies
EP0184187A2 (en) 1984-12-04 1986-06-11 Teijin Limited Mouse-human chimaeric immunoglobulin heavy chain, and chimaeric DNA encoding it
US4666829A (en) 1985-05-15 1987-05-19 University Of California Polypeptide marker for Alzheimer's disease and its use for diagnosis
JPS62100291A (en) 1985-10-28 1987-05-09 Teijin Ltd Gene fragment and plasmid
WO1987002671A1 (en) 1985-11-01 1987-05-07 International Genetic Engineering, Inc. Modular assembly of antibody genes, antibodies prepared thereby and use
US5547841A (en) 1987-10-08 1996-08-20 The Mclean Hospital Corporation In vitro method for screening for drugs that inhibit production or degradation of human A4-amyloid
US5134062A (en) 1988-03-22 1992-07-28 Cornell Research Foundation, Inc. Diagnosis of neuronal disorders and screening potential therapeutic agents therefor
EP0391714A2 (en) 1989-04-05 1990-10-10 BRIGHAM &amp; WOMEN'S HOSPITAL Diagnostic method for Alzheimer's disease: examination of non-neural tissue
WO1990012870A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Monoclonal antibody to amyloid peptide
WO1990012871A1 (en) 1989-04-14 1990-11-01 Research Foundation For Mental Hygiene, Inc. Cerebrovascular amyloid protein-specific monoclonal antibody sv17-6e10
US5234814A (en) 1989-06-01 1993-08-10 Du Pont Merck Pharmaceutical Company Diagnostic assay for alzheimer's disease
US5221607A (en) 1989-09-18 1993-06-22 Scios Nova Inc. Assays and reagents for amyloid deposition
WO1991004339A1 (en) 1989-09-18 1991-04-04 California Biotechnology Inc. Assays and reagents for amyloid deposition
EP0444856A2 (en) 1990-02-26 1991-09-04 Albert Einstein College Of Medicine Of Yeshiva University Diagnostic assay for Alzheimer's disease
WO1991016628A1 (en) 1990-04-24 1991-10-31 The Regents Of The University Of California Purification, detection and methods of use of protease nexin-2
WO1991019810A1 (en) 1990-06-15 1991-12-26 California Biotechnology Inc. Transgenic non-human mammal displaying the amyloid-forming pathology of alzheimer's disease
US5387742A (en) 1990-06-15 1995-02-07 Scios Nova Inc. Transgenic mice displaying the amyloid-forming pathology of alzheimer's disease
WO1992000521A1 (en) 1990-06-29 1992-01-09 Case Western Reserve University Diagnostic and prognostic methods based on soluble derivatives of the beta amyloid protein precursor
US5200339A (en) 1990-08-17 1993-04-06 Abraham Carmela R Proteases causing abnormal degradation of amyloid β-protein precursor
WO1992009699A1 (en) 1990-11-27 1992-06-11 L.F. Will & Cie (Belgique) Monoclonal antibody for use in diagnosing alzheimer's disease, hybridoma secreting said antibody and preparation method therefor
WO1992013069A1 (en) 1991-01-21 1992-08-06 Imperial College Of Science, Technology & Medicine Test and model for alzheimer's disease
WO1993001817A1 (en) 1991-07-25 1993-02-04 Charles Lwanga Ssali Antiviral compositions comprising fusidic acid, l-ascorbic acid and salicylic acid and derivatives
WO1993008264A1 (en) 1991-10-17 1993-04-29 Valio Bioproducts Ltd. Colostrum fraction, preparation and use thereof as cell culture media supplement
WO1993014200A1 (en) 1992-01-07 1993-07-22 Tsi Corporation Transgenic animal models for alzheimer's disease
WO1993016101A1 (en) 1992-02-05 1993-08-19 Athena Neurosciences, Inc. Beta-amyloid peptide production inhibitors and methods for their identification
US5538845A (en) 1992-02-05 1996-07-23 Athena Neurosciences, Inc. Beta-amyloid peptide production inhibitors and methods for their identification
US6018024A (en) 1992-04-15 2000-01-25 Elan Pharmaceuticals Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
US5721130A (en) 1992-04-15 1998-02-24 Athena Neurosciences, Inc. Antibodies and fragments thereof which bind the carboxyl-terminus of an amino-terminal fragment of βAPP
US5604102A (en) 1992-04-15 1997-02-18 Athena Neurosciences, Inc. Methods of screening for β-amyloid peptide production inhibitors
WO1993021526A1 (en) 1992-04-15 1993-10-28 Athena Neurosciences, Inc. METHODS AND COMPOSITIONS FOR MONITORING CELLULAR PROCESSING OF β-AMYLOID PRECURSOR PROTEIN
US5441870A (en) 1992-04-15 1995-08-15 Athena Neurosciences, Inc. Methods for monitoring cellular processing of β-amyloid precursor protein
US5455169A (en) 1992-06-04 1995-10-03 Alzheimer's Institute Of America, Inc. Nucleic acids for diagnosing and modeling Alzheimer's disease
WO1994000569A1 (en) 1992-06-18 1994-01-06 Genpharm International, Inc. Methods for producing transgenic non-human animals harboring a yeast artificial chromosome
US5593846A (en) 1992-07-10 1997-01-14 Athena Neurosciences Methods for the detection of soluble β-amyloid peptide
US5837672A (en) 1992-07-10 1998-11-17 Athena Neurosciences, Inc. Methods and compositions for the detection of soluble β-amyloid peptide
US5766846A (en) 1992-07-10 1998-06-16 Athena Neurosciences Methods of screening for compounds which inhibit soluble β-amyloid peptide production
WO1994001772A1 (en) 1992-07-13 1994-01-20 The Children's Medical Center Corporation SCREEN FOR ALZHEIMER'S DISEASE THERAPEUTICS BASED ON β-AMYLOID PRODUCTION
US5605811A (en) 1992-10-26 1997-02-25 Athena Neurosciences, Inc. Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
WO1994010569A1 (en) 1992-10-26 1994-05-11 Schenk Dale B METHODS AND COMPOSITIONS FOR THE DETECTION OF SOLUBLE β-AMYLOID PEPTIDE
US5750349A (en) 1993-01-25 1998-05-12 Takeda Chemical Industries Ltd. Antibodies to β-amyloids or their derivatives and use thereof
US5955317A (en) 1993-01-25 1999-09-21 Takeda Chemical Industries, Ltd. Antibodies to β-amyloids or their derivatives and use thereof
WO1994023049A2 (en) 1993-04-02 1994-10-13 The Johns Hopkins University The introduction and expression of large genomic sequences in transgenic animals
US6610493B1 (en) 1993-06-17 2003-08-26 Brigham And Women's Hospital Screening compounds for the ability to alter the production of amyloid-β peptide
US5612486A (en) 1993-10-27 1997-03-18 Athena Neurosciences, Inc. Transgenic animals harboring APP allele having swedish mutation
US5850003A (en) 1993-10-27 1998-12-15 Athena Neurosciences Transgenic rodents harboring APP allele having swedish mutation
WO1996015452A1 (en) 1994-11-14 1996-05-23 Athena Neurosciences, Inc. METHODS FOR AIDING IN THE DIAGNOSIS OF ALZHEIMER'S DISEASE BY MEASURING AMYLOID-β PEPTIDE (X- ≥41) AND TAU
WO1997010601A1 (en) 1995-09-14 1997-03-20 Samsung Electronics Co., Ltd. Refresh strategy for drams
WO1997048983A1 (en) 1996-06-18 1997-12-24 Athena Neurosciences, Inc. SCREENING COMPOUNDS FOR THE ABILITY TO ALTER THE PRODUCTION OF AMYLOID-β PEPTIDE (x-≥41)

Non-Patent Citations (261)

* Cited by examiner, † Cited by third party
Title
"Alzheimer's Assault," ScienceScope, p. 1059 (Feb. 28, 1992).
Abraham et al., "A Calcium-activated Protease from Alzheimer's DiseaseBrain Cleaves at the N-terminus of the Amyloid beta-protein" Biochem. Biophys. Res. Comm., 174(2):790-796 (1991).
Abraham et al., "A Calcium-activated Protease from Alzheimer's DiseaseBrain Cleaves at the N-terminus of the Amyloid β-protein" Biochem. Biophys. Res. Comm., 174(2):790-796 (1991).
Ali et al., "More Transgenic Mouse Studies of Alzheimer Amyloid Precursor (AAP) Proteins and Derivatives," Society for Neuroscience Abstracts, 18(2):Abstract 616.8, 22nd Annual Meeting, Anaheim, CA Oct. 25-30, 1992.
Allison et al., "Diabetes in transgenic mice resulting from over-expression of class I histocompatibility molecules in pancreatic beta cells," Nature, 333:529-533 (1988).
Allison et al., "Diabetes in transgenic mice resulting from over-expression of class I histocompatibility molecules in pancreatic β cells," Nature, 333:529-533 (1988).
Allsop et al., "Immunohistochemical Evidence for the Derivative of a Peptide Ligand from the Amyloid .beta.-Protein Precursor of Alzheimer Disease," PNAS, 85:2790-2794 (1988).
Anderson et al., "Exact Cleavage Site of Alzheimer Amyloid Precursor in Neuronal PC-12 Cells," Neuroscience Letters, 128:126-128 (1991).
Antal et al., "Animal Models of Alzheimer's, Parkinson's and Huntington's Disease. A Minireview," Neurobiology, 1(2):101-122 (1993).
Asami-Odaka et al. "Long Amyloid β-Protein Secreted Wild-Type Human Neuroblastoma IMR-32 Cells", Biochemistry 34:10272-10278 (1995).
Asami-Odaka et al., "Long amyloid beta-protein secreted from wild-type human neuroblastoma IMR-32 cells," Biochemistry, 34(32):10272-10278 (1995).
Barrow et al., "Solution structures of beta peptide and its constituent fragments: relation to amyloid deposition," Science, 253:179-182 (1991).
Boller et al., Biological Markers of Alzheimer's Disease, Springer-Verlag (Berlin), pp. 24-29 (1989).
Bullock et al., Techniques in Diagnostic Pathology, vol. 2, pp. 100-112, Academic Press (London), (1991).
Burdick et al., "Assembly and aggregation of synthetic Alzheimer's A4/beta amyloid peptide analogs," J. Biol. Chem., 267(1):546-554 (1992).
Buxbaum et al. "Protein phosphorylation inhibits production of Alzheimer amyloid beta/A4 peptide," PNAS, 90:9195-9198 (1993).
Cai et al., "Release of Excess Amyloid beta Protein from a Mutant Amyloid Protein Precursor," Science, 259:514-516 (1993).
Cai et al., "Release of Excess Amyloid β Protein from a Mutant Amyloid Protein Precursor," Science, 259:514-516 (1993).
Carroll et al., "An Age-Related Correlation Between Levels of .beta.-Amyloid Precursor Protein and .beta.-Amyloid in Human Cerebrospinal Fluid," Biochem. Biophys. Res. Comm., 210(2):345-349 (1995).
Castano et al., "In Vitro Formation of Amyloid Fibrils from Two Synthetic Peptides of Different Lengths Homologous to Alzheimer's Disease .beta. Protein," Biochem. Biophys. Res. Comm., 141(2):702-789 (1986).
Ceballos-Picot et al., "Neuronal-specific expression of human copper-zinc superoxide dismutase gene in transgenic mice: animal model of gene dosage effects in Down's syndrome," Brain Research, 552:198-214 (1991).
Chartier-Harlan et al., "Early-onset Alzheimer's disease caused by mutations at codon 717 of the beta-amyloid precursor protein gene," Nature, 353:844-846 (1991).
Chartier-Harlan et al., "Early-onset Alzheimer's disease caused by mutations at codon 717 of the β-amyloid precursor protein gene," Nature, 353:844-846 (1991).
Checler et al. "Processing of the β-Amyloid Precursor Protein and Its Regulation in Alzheimer's Disease", J. Neurochemistry 65:1431-1444 (1995).
Chiu et al., Archive of Neurology, 50(1):57-63 (1993).
Citron et al., "Mutation of the beta-amyloid precursor protein in familial Alzheimer's disease increases beta-protein production," Nature, 360:672-674 (1992).
Citron et al., "Mutation of the β-amyloid precursor protein in familial Alzheimer's disease increases β-protein production," Nature, 360:672-674 (1992).
Cotton, R.G.H., "A G to C Transversion in Codon 258 of the alpha-Subunit of beta-Hexosaminidase A in an Infant Tay-Sachs Disease Patient," Human Mutation, 2:496-497 (1993).
Cotton, R.G.H., "A G to C Transversion in Codon 258 of the α-Subunit of β-Hexosaminidase A in an Infant Tay-Sachs Disease Patient," Human Mutation, 2:496-497 (1993).
Crawford et al., "Alzheimer's Disease Untangled," BioEssays, 14(11):727-734 (1992).
De Strooper et al., "Study of the Synthesis and Secretion of Normal and Artificial Mutants of Murine Amyloid Precursor Protein (APP): Cleavage of APP Occurs in a Late Compartment of the Default Secretion Pathway," J. Cell Biology, 121(2):295-304 (1993).
Dovey et al., "Cells with a familial Alzheimer's disease mutation produce authentic beta-peptide," NeuroReport, 4:1039-1042 (1993).
Dovey et al., "Cells with a familial Alzheimer's disease mutation produce authentic β-peptide," NeuroReport, 4:1039-1042 (1993).
Dyrks et al., "Generation of .beta. A4 from the amyloid protein precursor and fragments thereof," FEBS Letters, 335(1):89-103 (1993).
EP 0 906 577, Notice of Opposition (English & German versions) filed by Boehringer Ingelheim on Jun. 3, 2009.
Epstein et al., "Transgenic mice with increased Cu/Zn-superoxide dismutase activity: Animal model of dosage effects in Down syndrome," PNAS, 84:8044-8048 (1987).
Erickson, D., "Model Mice, Transgenic animals aid Alzheimer's research," p. 34, Scientific American, (1991).
Esch et al., "Cleavage of amyloid beta peptide during constitutive processing of its precursor," Science, 248:1122-1124 (1990).
Esch et al., "Cleavage of amyloid β peptide during constitutive processing of its precursor," Science, 248:1122-1124 (1990).
Estus et al., "Potentially Amyloidogenic, Carboxyl-Terminal Derivatives of the Amyloid Protein Precursor," Science, 255:726-728 (1992).
Felsenstein et al., "Transgenic Rat and In-Vitro Studies of beta-Amyloid Precursor Protein Processing," pp. 401-409 from Alzheimer's and Parkinson's Diseases, edited by Hanin, I. et al., Plenum Press, New York, (1995).
Felsenstein et al., "Transgenic Rat and In-Vitro Studies of β-Amyloid Precursor Protein Processing," pp. 401-409 from Alzheimer's and Parkinson's Diseases, edited by Hanin, I. et al., Plenum Press, New York, (1995).
Fidani et al., "Screening for mutations in the open reading frame and promoter of the beta-amyloid precursor protein gene in familial Alzheimer's disease: identification of a further family with APP717 Val Ile," Human Molecular Genetics, 1(3):165-168 (1992).
Fidani et al., "Screening for mutations in the open reading frame and promoter of the β-amyloid precursor protein gene in familial Alzheimer's disease: identification of a further family with APP717 Val Ile," Human Molecular Genetics, 1(3):165-168 (1992).
Fisher et al., "Expression of the amyloid precursor protein gene in mouse oocytes and embryos," PNAS, 88:1779-1782 (1991).
Forss-Petter et al., "Transgenic Mice Expressing beta-Galactosidase in Mature Neurons under Neuron-Specific Enolase Promoter Control," Neuron, 5:187-197 (1990).
Forss-Petter et al., "Transgenic Mice Expressing β-Galactosidase in Mature Neurons under Neuron-Specific Enolase Promoter Control," Neuron, 5:187-197 (1990).
Francis et al., "Animal and Drug Modelling for Alzheimer Synaptic Pathology," Progress in Neurobiology, 39:517-545 (1992).
Fraser et al., "Biochemistry of Alzheimer's Disease Amyloid Plaques," Clin. Biochem., 26:339-349 (1993).
Fukuchi et al., "Intestinal beta-Amyloidosis in Transgenic Mice," abstract 421.16, Society for Neuroscience Abstracts, 19:1035 (1993).
Fukuchi et al., "Intestinal β-Amyloidosis in Transgenic Mice," abstract 421.16, Society for Neuroscience Abstracts, 19:1035 (1993).
Fukuchi et al., "Transgenic Animal Models for Alzheimer's Disease," Ann. NY Acad. Sci., 695:217-223 (1993).
Gallagher et al., "Animal models of normal aging: relationship between cognitive decline and markers in hippocampal circuitry," Behavioural Brain Research, 57:155-162 (1993).
Games et al., "Alzheimer-type neuropathology in transgenic mice overexpressing V717F beta-amyloid precursor," Nature, 373:523-527 (1995).
Gene Targeting A Practical Approach, edited by Joyner, A.L., Oxford Univ. Press (1993) cover page, colophon, and table of contents.
Glenner et al., "Alzheimer's disease and Down's Syndrome: Sharing of unique cerebrovascular amyloid fibril protein," Biochem. Biophys. Res. Comm., 122:1131-1135 (1984).
Glenner et al., "Alzheimer's disease: Initial report of the purification and characterization of a novel cerebrovascular amyloid protein," Biochem. Biophys. Res. Comm., 120:885-890 (1984).
Goate et al., "Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease," Nature, 349:704-706 (1991).
Goding, James W., "Production and Application of Monoclonal Antibodies in Cell Biology, Biochemistry and Immunology," in: Monoclonal Antibodies: Principles and Practice, Ch. 3, pp. 72-74, Academic Press, London (1983).
Golde et al., "Processing of the Amyloid Protein Precursor to Potentially Amyloidgenic Derivatives," Science, 255:728-730 (1992).
Golde et al., "Production of Amyloid beta Protein from Normal Amyloid beta-Protein Precursor (betaAPP) and the Mutated betaAPPS Linked to Familial Alzheimer's Disease," from Alzheimer's Disease Amyloid Precursors Proteins, Signal Transduction, and Neural Transplantation, 695:103-108, by Ann. New York Acad. Sci. (1993).
Golde et al., "Production of Amyloid β Protein from Normal Amyloid β-Protein Precursor (βAPP) and the Mutated βAPPS Linked to Familial Alzheimer's Disease," from Alzheimer's Disease Amyloid Precursors Proteins, Signal Transduction, and Neural Transplantation, 695:103-108, by Ann. New York Acad. Sci. (1993).
Goldgaber et al., "Characterization and Chromosomal Localization of a cDNA Encoding Brain Amyloid of Alzheimer's Disease," Science, 235:877-880 (1987).
Goverman et al., "Transgenic Mice That Express a Myelin Basic Protein-Specific T Cell Receptor Develop Spontaneous Autoimmunity," Cell, 72:551-560 (1993).
Gravina et al., "Amyloid beta protein (A beta) in Alzheimer's disease brain. Biochemical and immunocytochemical analysis with antibodies specific for forms ending at A beta 40 or A beta 42(43)," J. Biol. Chem., 270:7013-7016 (1995).
Greaves et al., "A transgenic mouse model of sickle cell disorder," Nature, 343:183-185 (1990).
Greenberg et al., "Transgenic Mouse Studies of Alzheimer Amyloid Precursor (APP) Proteins and Deravitives," Society for Neuroscience Abstracts, 18(part 2), abstract 616.7 (1992).
Greenberg et al., "Yet More Transgenic Mouse Studies of Alzheimer Amyloid Precursor (APP)," Soc. for Neurosci. Abstracts, 19:1035, abst. 421.12 (1993).
Haass et al., "Amyloid beta-peptide is produced by cultured cells during normal metabolism," Nature, 359:322-325 (1992).
Haass et al., "Amyloid β-peptide is produced by cultured cells during normal metabolism," Nature, 359:322-325 (1992).
Haass et al., "beta-Amyloid peptide and a 3-kDa fragment are derived by distinct cellular mechanisms," J. Biol. Chem., 268(5):3021-3024 (1993).
Haass et al., "Cellular Processing of beta-Amyloid Precursor Protein and the Genesis of Amyloid beta-Peptide," Cell, 75:1039-1042 (1993).
Haass et al., "Cellular Processing of β-Amyloid Precursor Protein and the Genesis of Amyloid β-Peptide," Cell, 75:1039-1042 (1993).
Haass et al., "Mutations associated with a locus for familial Alzheimer's disease result in alternative processing of amyloid beta-protein precursor," J. Biol. Chem., 269:17741-17748 (1994).
Haass et al., "The Swedish mutation causes early-onset Alzheimer's Disease by beta-secretase cleavage within the secretory pathway," Nature Med., 1:1291-1296 (1995).
Haass et al., "β-Amyloid peptide and a 3-kDa fragment are derived by distinct cellular mechanisms," J. Biol. Chem., 268(5):3021-3024 (1993).
Hammer et al., "Partial correction of murine hereditary growth disorder by germ-line incorporation of a new gene," Nature, 311:65-67 (1984).
Hardy, J., "Framing beta-amyloid," Nature Genetics, 1:233-234 (1992).
Hardy, J., "Framing β-amyloid," Nature Genetics, 1:233-234 (1992).
Hardy, J., "The Alzheimer family of diseases: Many etiologies, one pathogenesis?," PNAS, 94:2095-2097 (1997).
Harlow et al., Antibodies A Laboratory Manual, (Cold Spring harbor Laboratory), pp. 578-582 (1988).
Hendricks et al., "Presenile dementia and cerebral haemorrhage linked to a mutation at codon 692 of the beta-amyloid precursor protein gene," Nature Genetics, 1:218-221 (1992).
Hendricks et al., "Presenile dementia and cerebral haemorrhage linked to a mutation at codon 692 of the β-amyloid precursor protein gene," Nature Genetics, 1:218-221 (1992).
Hendriksson et al., "Analysis and quantitation of the beta-amyloid precursor protein in the cerebrospinal fluid of Alzheimer's disease patients with a monoclonal antibody-based immunoassay," J. Neurochem., 56:1037 (1991).
Higaki et al., "Inhibition of beta-amyloid formation identifies proteolytic precursors and subcellular site of catabolism," Neuron, 14:651-659 (1995).
Higgins et al., "Transgenic Mice Expressing Human beta-APP751, But Not Mice Expressing beta-APP695, Display Early Alzheimer's Disease-like Histophathology," Annals NY Acad. Sci., 695:224-227 (1993).
Higgins et al., "Transgenic Mice Expressing Human β-APP751, But Not Mice Expressing β-APP695, Display Early Alzheimer's Disease-like Histophathology," Annals NY Acad. Sci., 695:224-227 (1993).
Higgins et al., "Transgenic Mouse Brain Histopathology Resembles Early Alzheimer's Disease " Ann. Neurol., 35:598-607 (1994).
Hilbich et al., "Aggregation and secondary structure of synthetic amyloid beta A4 peptides of Alzheimer's disease," J. Mol. Biol., 218:149-163 (1991).
Hogan et al., Manipulating the Mouse Embryo, A Laboratory Manual, Cold Spring Harbor Laboratory, (1986) cover page, colophon, and table of contents.
Holtzman et al., "Molecular studies in Alzheimer's disease," TIBS, 16:140-144 (1991).
Howland et al., "Neuron-Specific Expression of Human Beta-Amyloid Precursor Protein (APP) in Transgenic Mice," Society for Neuroscience Abstracts, 19:1035, abstract 421.13 (1993).
Hsiao et al., "Spontaneous Neurodegeneration in Transgenic Mice with Mutant Prion Protein," Science, 250:1587-1590 (1990).
Hung et al., "Activation of protein kinase C inhibits cellular production of the amyloid beta-protein," J. Biol. Chem., 268:22959-22962 (1993).
Hyman et al., "Amyloid, dementia and Alzheimer's disease," Curr. Opin. Neurology Neurosurgery, 5:88-93 (1992).
Hyman et al., "Kunitz Protease Inhibitor-Containing Amyloid beta-protein Precursor Immunoreactivity in Alzheimer's Disease " J. Neuropath. Exp. Neurol., 51:76-83 (1992).
Hyman et al., "Kunitz Protease Inhibitor-Containing Amyloid β-protein Precursor Immunoreactivity in Alzheimer's Disease " J. Neuropath. Exp. Neurol., 51:76-83 (1992).
Iwamoto et al., "Neuroblastoma in a transgenic mouse carrying a metallothionein/ret fusion gene," Br. J. Cancer, 67:504-507 (1993).
Iwatsubo et al., "Amyloid beta protein (A beta) deposition: A beta 42(43) precedes A beta 40 in Down Syndrome," Ann. Neurol., 37(3):294-299 (1995).
Iwatsubo et al., "Visualization of A beta 42(43) and A beta 40 in senile plaques with end-specific A beta monoclonals: evidence that an initially deposited species is A beta 42(43)," Neuron, 13(1):45-53 (1994).
Jan et al., "Receptor-regulated ion channels," Curr. Opin. Cell Biology, 9:155-160 (1997).
Jarrett et al., "Models of the β Protein C-Terminus: Differences in Amyloid Structure May Lead to Segregation of "Long" and "Short" Fibrils" J. Am. Chem. Soc. 116: 9741-9742 (1994).
Jarrett et al., "The carboxy terminus of the beta amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer's disease," Biochemistry 32(18):4693-4697 (1993).
Jarrett et al., "The C-Terminus of the β Protein is Critical in Amyloidogenesis", Annals of the New York Academy of Sciences 695: 144-148 (1993).
Joachim et al., "Amyloid beta-protein deposition in tissues other than brain in Alzheimer's disease," Nature, 341:226 (1989).
Joachim et al., "Protein chemical and immunocytochemical studies of meningovascular beta-amyloid protein in Alzheimer's disease and normal aging," Brain Research, 474(1):100-111 (1988).
Jones et al., "Mutation in codon 713 of the beta amyloid precursor protein gene presenting with schizophrenia," Nature Genetics, 1:306-309 (1992).
Jones et al., "Mutation in codon 713 of the β amyloid precursor protein gene presenting with schizophrenia," Nature Genetics, 1:306-309 (1992).
Kalaria et al., "Abundance of the longer Aβ42 in neocortical and cerbrovascular Amyloid β deposits in Swedish familial Alzheimer's disease and Down's syndrome", NeuroReport 7:1377-1381 (1996).
Kammesheidt et al., "Deposition of beta/A4 immunoreactivity and neural pathology in transgenic mice expressing the carboxyl-terminal fragment of the Alzheimer amyloid precursor in the brain," PNAS, 89:10857-10861 (1992).
Kammesheidt et al., "Deposition of β/A4 immunoreactivity and neural pathology in transgenic mice expressing the carboxyl-terminal fragment of the Alzheimer amyloid precursor in the brain," PNAS, 89:10857-10861 (1992).
Kang et al., "The precursor of Alzheimer's disease amyloid A4 protein resembles a cell-surface receptor," Nature, 325:733-736 (1987).
Kawarabayashi et al., "Accumulation of β-Amyloid Fibrils in Pancreas of Transgenic Mice", Neurobiol. of Aging 17(2): 215-222 (1996).
Keffer et al., "Transgenic mice expressing human tumour necrosis factor: a predictive genetic model of arthritis," EMBO J., 10(13):4025-4031 (1991).
Kennedy et al., "Familial Alzheimer's disease," Brain, 116:309-324 (1993).
Kennedy et al., "Only Kunitz-Inhibitor-Containing Isoforms of Secreted Alzheimer Amyloid Precursor Protein Show Amyloid Immunoreactivity in Normal Cerebrospinal Fluid," Neurodegeneration, 1:59-64 (1992).
Kim et al., "Detection and Quantitation of Amyloid B-Peptide with 2 Monoclonal Antibodies," Neuroscience Research Communications, 7(2):113-122 (1990).
Kirschner et al., "Synthetic Peptide Homologous to .beta. Protein From Alzheimer Disease Forms Amyloid-Like Fibrils in vitro," PNAS, 84:6953-6957 (1987).
Kitaguchi et al., "Novel precursor of Alzheimer's disease amyloid protein shows protease inhibitory activity," Nature, 331:530-532 (1988).
Knops et al., "Cell-type and amyloid precursor protein-type specific inhibition of A beta release by bafilomycin A1, a selective inhibitor of vacuolar ATPases," J. Biol. Chem. 270(6):2419-2422 (1995).
Koliatsos et al., "Neurotrophic Strategies for Treating Alzheimer's Disease: Lessons from Basic Neurobiology and Animal Models," from Alzheimer's Disease Amyloid Precursors Proteins, Signal Transduction, and Neural Transplantation, vol. 695, pp. 292-299, by Ann. NY Acad. Sci., (1993).
Konig et al., "Identification and Differential Expression of a Novel Alternative Splice Isoform of the betaA4 Amyloid Precursor Protein (APP) mRNA in Leukocytes and Brain Microglial Cells " J. Biol. Chem., 267(15):10804-10809 (1992).
Konig et al., "Identification and Differential Expression of a Novel Alternative Splice Isoform of the βA4 Amyloid Precursor Protein (APP) mRNA in Leukocytes and Brain Microglial Cells " J. Biol. Chem., 267(15):10804-10809 (1992).
Koo et al., "Evidence that production and release of amyloid beta-protein involves the endocytic pathway," J. Biol. Chem. 269(26):17386-17389 (1994).
Korf et al., "S-Antigen and Rod-Opsin Immunoreactions in Midline Brain Neoplasms of Transgenic Mice: Similarities to Pineal Cell Tumors and Certain Medulloblastomas in Man," J. Neuropath. Exper. Neurology, 49(4):424-437 (1990).
Kozak, M., "The Scanning Model for Translation: An Update," J. Cell Biology, 108:229-241 (1989).
Kozlowski et al., "The Neurotoxic Carboxy-Terminal Fragment of the Alzheimer Amyloid Precursor Binds Specificity to a Neuronal Cell Surface Molecule: pH Dependance of the Neurotoxicity and the Binding," J. Neuroscience, 12(5):1679-1687 (1992).
Ksiezak-Reding et al., "Alz 50, a monoclonal antibody to Alzheimer's disease antigen, cross reacts with tau proteins from bovine and normal human brain," J. Biol. Chem., 263(17):7943-7947 (1988).
Lamb et al., "Introduction and expression of the 400 kilobase precursor amyloid protein gene in transgenic mice," Nature Genetics, 5:22-30 (1993).
Lannfelt et al., "Alzheimer's disease: molecular genetics and transgenic animal models," Behav. Brain Research, 57(2):207-213 (1993).
Lannfelt et al., "Low frequency of the APP 670/671 mutation in familial Alzheimer's disease in Sweden," Neuroscience Letters, 153:85-87 (1993).
Lavigueur et al., "High Incidence of Lung, Bone, and Lymphoid Tumors in Transgenic Mice Overexpressing Mutant Alleles of the p53 Oncogene " Mol. Cellular Biol., 9(9):3982-3991 (1989).
Lee et al., "A68: a major subunit of paired helical filaments and derivatized forms of normal Tau," Science, 251:675-678 (1991).
Lemere et al., "Sequence of deposition of heterogeneous amyloid beta-peptides and APO E in Down Syndrome: implications for initial events in amyloid plaque formation," Neurobiol. Dis., 3(1):16-32 (1996).
Levy et al., "Mutation of the Alzheimer's Disease Amyloid Gene in Hereditary Cerebral Hemorrhage, Dutch Type," Science, 248:1124-1126 (1990).
Lieberburg et al., "Expression of Human Alzheimer's Amyloid Precursor Protein in Transgenic Mice," Soc. Neuroscience Abstracts, vol. 19, abstract 421.15 (1993).
Luo et al., "Human Amyloid Precursor Protein Ameliorates Behavior Deficit of Flies Deleted for Appl Gene," Neuron, 9:595-605 (1992).
Mak, et al., "Polyclonals to β-amyloid (1-42) identify most plaque and vascular deposits in Alzheimer cortex, but not striatum," Brain Res., 667:138-142 (1994).
Mann et al., "Microglial cells and Amyloid β protein (Aβ) deposition: association with Aβ40-containing plaques", Acta Neuropathol. 90: 472-477 (1995).
Mann et al., "Predominant deposition of amyloid-beta 42(43) in plaques in cases of Alzheimer's disease and hereditary cerebral hemorrhage associated with mutatuibs in the amyloid precursor protirn gene," Amer. J. Pathol. APR, 4(148): 1257-1266 (1996).
Marx, J., "Alzheimer's Research Moves to Mice," Science, 253:266-267 (1991).
Marx, J., "Major Setback for Alzheimer's Models," Science, 255:1200-1202 (1992).
Marx, J., "New Lead to an Alzheimer's Mouse?," Science, 261:1520 (1993).
Masters et al., "Amyloid plaque core protein in Alzheimer disease and Down Syndrome," PNAS, 82(12):4245-4249 (1985).
McKhann et al., "Clinical Diagnosis of Alzheimer's Disease," Neurology, 34:939-944 (1984).
Mehta et al., Techniques in Diagnostic Pathology, eds. Bullock et al., Academic Press, Boston, p. 106.
Miller et al., "Alzheimer's disease: transgenic models to test new chemicals and pharmaceuticals," Curr: Opin. Biotechnology, 3:683-686 (1992).
Moran et al., "Age-related learning deficits in transgenic mice expressing the 751-amino acid isoform of human β-amyloid precursor protein," PNAS, 92:5341-5345 (1995).
Mullan et al., "A pathenogenic mutation for probable Alzheimer's disease in the APP gene at the N-terminus of β-amyloid," Nature Genetics, 1:345-347 (1992).
Mullan et al., "Genetic and molecular advances in Alzheimer's disease," TINS, 16(10):398-403 (1993).
Mullan, M., "Familial Alzheimer's disease: second gene locus located," BMJ, 305:1108-1109 (1992).
Mullins et al., "Fulminant hypertension in transgenic rats harbouring the mouse Ren-2 gene," Nature, 344:541-544 (1990).
Murrell et al., "A Mutation in the Amyloid Precursor Protein Associated with Hereditary Alzheimer's Disease " Science, 254:97-99 (1991).
Nakamura et al., "Amyloid .beta. Protein Levels in Cerebrospinal Fluid Are Elevated in Early-onset Alzheimer's Disease," Annals of Neurology, 36(6):903-911 (1994).
Neve et al., "Brain transplants of cells expressing the carboxyl-terminal fragment of the Alzheimer amyloid protein precursor cause specific neuropathology in vivo," PNAS, 89:3448-3452 (1992).
Nussbaum et al., "Alzheimer's Disease and Amyloid Protein—in (Transgenic) Mice and Men," Harefuah, 123(9):362-364, document in Hebrew (1992).
Oltersdorf et al., "The secreted form of the Alzheimer's amyloid precursor protein with the Kunitz domain is protease nexin-II," Nature, 341:144-147 (1989).
Order Denying Mayo's Ex Patre Motion to Stay Deadline for Motion for Attorney's Fees, U.S. District Court for the Northern District of California, Case No. C99-04464 WHA.
Order Granting Defendant's Motion for Summary Judgment of Anticipation, U.S. District Court for the Northern District of California, Case No. C99-04464 WHA.
Palmert et al., "Soluable derivatives of the beta amyloid protein precursor in cerebrospinal fluid: alterations in normal aging and in Alzheimer's disease," Neurology, 40:1028-1034 (1990).
Palmert et al., "Soluble Derivatives of the β Amyloid Protein Precursor of Alzheimer's Disease are Labeled by Antisera to the β amyloid Protein," Biochem. Biophys. Res. Comm., 165(1):182-188 (1989).
Palmert et al., "The β-amyloid protein precursor of Alzheimer's disease has soluble derivatives found in human brain and cerebrospinal fluid," PNAS, USA 86:6338-6342 (1989).
Palmiter et al., "Dramatic growth of mice that develop from eggs microinjected with metallothionein-growth hormone fusion genes," Nature, 300:611-615 (1982).
Pardridge et al., "High molecular weight Alzheimer's disease amyloid peptide immunoreactivity in human serum and CSF is an immunoglobulin G," Biochem. Biophys. Res. Comm., 145(1):241-248 (1987).
Pavlin, R., "Brain Amyloid in Alzheimer's Disease—A New Experimental Model," Neurologia Croatica, 41(4):227-234 (1992).
PCT Search Report of 112/16/1993 for application PCT/US93/08264.
PCT Search Report of Nov. 27, 1997 for application PCT/US97/10601.
PCT Written Opinion of Jun. 22, 1994 for application PCT/US93/08264.
Pearson et al., "Expresion of the human β-amyloid precursor protein gene from a yeast artificial chromosome in transgenic mice," PNAS, 90:10578-10582 (1993).
Pennisi, "A Molecular Whodunit," Science News, 145(1):8-11 (1994).
Perraud et al., "The promoter of the human cystic fibrosis transmembrane conductance regulator gene directing SV40 T antigen expression induces malignant proliferation of ependymal cells in transgenic mice," Oncogene, 7:993-997 (1992).
Pirttila et al., "Soluble amyloid β-Protein in the cerebrospinal fluid from patients with Alzheimer's disease. Vascular dementia and controls," Journal of the Neurological Sciences, 127:90-95 (1994).
Podlisny et al., "Detection of soluble forms of the beta-amyloid precursor protein in human plasma," Biochem. Biophys. Res. Comm., 167(3):1094-1101 (1990).
Podlisny et al., "Homology of the amyloid beta protein precursor in monkey and human supports a primate model for beta amyloidosis in Alxheimer's disease," Am J. Pathol., 138(6):1423-1435 (1991).
Ponte et al., "A new A4 amyloid mRNA contains a domain homologous to serine proteinase inhibitors," Nature, 331:525-527 (1988).
Prayson et al., "The search for diagnostic criteria in Alzheimer's disease: an update," Cleveland Clinic Journal of Medicine, 61:115-122 (1994).
Price et al., "Alzheimer disease and the prion disorders amyloid .beta.-protein and Prion protein amyloidoses," PNAS, 90:6381-6384 (1993).
Price et al., "Alzheimer's Disease-Type Brain Abnormalities in Animal Models," Down Syndrome and Alzheimer Disease, pp. 271-287, Wiley-Liss, Inc., (1992).
Price et al., "Cellular and molecular biology of Alzheimer's disease and animal models," Annual Reviews, 45:435-446 (1994).
Pulliam et al., "Use of Aggregating Brain Cultures to Study the Replication of Herpes Simplex Virus Types 1 and 2 in Central Nervous System Tissue," J. Virol. Met., 9:301-316 (1984).
Quon et al., "Formation of β-amyloid protein deposits in brains of transgenic mice," Nature, 352:239-241 (1991).
Robakis et al., "An alternative secretase cleavage produces soluble Alzheimer amyloid precursor protein containing a potentially amyloidogenic sequence," Soc. Neurosci., Abstract No. 15.5, 22nd Annual Meeting, Anaheim, CA.
Robakis et al., "Molecular cloning and characterization of a cDNA encoding the cerebrovascular and the neuritic plaque amyloid peptides," PNAS, 84:4190-4194 (1987).
Roch et al., "Biologically Active Domain of the Scereted Form of the Amyloid β/A4 Protein Precursor," from Alzheimer's Disease Amyloid Precursor Proteins, Signal Transduction, and Neuronal Transplantation, Ann. NY Acad. Sci., 695:149-157 (1993).
Roher et al., "beta-Amyloid-(1-42) is a major component of cerebrovascular amyloid deposits: implications for the pathology of Alzheimer disease," PNAS 90(22):10836-10840 (1993).
Rumble et al., "Amyloid A4 protein and its precursor in Down's syndrome and Alzheimer's disease," N. Engl. J. Med., 320(22):1446-1452 (1989).
Ryan et al., "Human Sickle Hemoglobin in Transgenic Mice," Science, 247:566-568 (1990).
Sahasrabudhe et al., "Release of Amino-terminal Fragments from Amyloid Precursor Protein Reporter and Mutated Derivatives in Cultured Cells," J. Biol. Chemistry, 267(35):25602-25608 (1992).
Saido et al., "Spatial resolution of the primary beta-amyloidogenic process induced in postischemic hippocampus," J. Biol. Chem., 269(21):15253-15257 (1994).
Sandhu et al., "Expression of the Human β-Amyloid Protein of Alzheimer's Disease Specifically in the Brains of Transgenic Mice," J. Biol. Chemistry, 266(32):21331-21334 (1991).
Sarvetnick et al., "Insulin-Dependant Diabetes Mellitus Induced in Transgenic Mice by Ectopic Expression of Class II MHC and Interferon-Gamma," Cell, 52:773-782 (1988).
Schenk et al., "Therapeutic approaches related to amyloid-beta peptide and Alzheimer's disease," J. Med. Chem., 38(21):4141-4154 (1995).
Scheuner et al. (1995). Neurosci. Abstracts in press.
Scheuner et al., "Secreted amyloid beta-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease," Nature Medicine, 2(8):864-870 (1996).
Schlossmacher et al., "Detection of distinct isoform patterns of the beta-amyloid precursor protein in human platelets and lymphocytes," Neurobiol. Aging, 13(3):421-434 (1992).
Schwartz et al., "Human Amyloid Precursor Protein Expression in Transgenic Mice as a Model of Alzheimer's Disease: Search for Pathology," abstract 421.14, Society for Neuroscience Abstracts, 19:1035 (1993).
Scott et al., "Inability to Detect β-Amyloid Protein Precursor mRNA in Alzheimer Plaque-Assoicated Microglia," Experimental Neurology, 121:113-118 (1993).
Scott et al., "The Processing of Native and Mutant APP751 in Human 293 Cells," Neurobiology of Aging, 13(Supp. 1):578-579, abstract 310 (1992).
Scott et al., "Transgenic Mice Expressing Hamster Prion Protein Produce Species-Specific Scrapie Infectivity and Amyloid Plaques," Cell, 59:847-857 (1989).
Selkoe et al., "Altered structural proteins in plaques and tangles: what do they tell us about the biology of Alzheimer's disease," Neurobiol. Aging, 7(6):425-432 (1986).
Selkoe et al., "Isolation of low-molecular-weight proteins from amyloid plaque fibers in Alzheimer's disease," J. Neurochem., 46(6):1820-1834 (1986).
Selkoe et al., "Molecular pathology of amyloidogenic proteins and the role of vascular amyloidosis in Alzheimer's disease," Neurobiol. Aging, 10(5):387-395 (1989).
Selkoe et al., "Physiological production of the β-amyloid protein and the mechanism of Alzheimer's disease," Trends in Neuroscience, 16(10):403-409 (1993).
Selkoe et al., "β-amyloid precursor protein of Alzheimer disease occurs as 110- to 135-kilodalton membrane-associated proteins in neural and nonneural tissues," PNAS, 85:7341-7345 (1988).
Selkoe, "Alzheimer's disease: a central role for amyloid," J. Neuropathol. Exp. Neurol., 53(5):438-447 (1994).
Selkoe, "The molecular pathology of Alzheimer's disease," Neuron 6(4):487-498 (1991).
Seubert et al., "Isolation and quantification of soluble Alzheimer's β-peptide from biological fluids," Nature, 359:325-327 (1992).
Seubert et al., "Secretion of β-amyloid precursor protein cleaved at the amino terminus of the β amyloid peptide," Nature, 361:260-263 (1993).
Shoji et al., "Production of the Alzheimer Amyloid β Protein by Normal Proteolytic Processing," Science, 258:126-129 (1992).
Siman et al., "Processing of the β-Amyloid Precursor Multiple Proteases Generate and Degrade Potentially Amyloidogenic Fragments," J. Biol. Chemistry, 268(22):16602-16609 (1993).
Sisodia, S.S., "β-Amyloid precursor protein cleavage by a membrane-bound protease," PNAS, 6075-6079 (1992).
Snyder et al., "Amyloid β Aggregation: Selective Inhibition of Aggregation in Mixtures of Amyloid with Different Chain Lengths", Biophys. J. 67: 1216-1228 (1994).
Sofroniew et al., "Transgenic modelling of neurodegenerative events gathers momentum," TINS, 14(12):513-514 (1991).
Stacey et al., "Perinatal lethal osteogenesis imperfecta in transgenic mice bearing an engineered mutant pro-α1(I) collagen gene," Nature, 332:131-136 (1988).
Stout et al., "Expression of human HPRT in the central nervous system of transgenic mice," Nature, 317:250-253 (1985).
Suzuki et al., "An increased percentage of long amyloid beta protein secreted by familial amyloid beta protein precursor (beta APP717) mutants," Science 264:1336-1340 (1994).
Suzuki et al., "High Tissue Content of Soluble β1-40 is Linked to Cerebral Amyloid Angiopathy", Amer. J. Pathol. 145(2): 452-460 (1994).
Tabaton et al., "Soluble Amyloid β Protein is a Marker of Alzheimer Amyloid in the Brain But Not in Cerebrospinal Fluid," Neurology, 44(suppl 2):A371 (1994).
Tamaoka et al. "APP717 Missense Mutation Affects the Ratio of Amyloid β Protein Species (Aβ1-42/43 and Aβ1-40) in Familial Alzheimer's Disease Brain", J. Biol. Chem. 269(52): 32721-32724 (1994).
Tamaoka et al., "Amyloid β protein 1-42/43 (Aβ1-42/43) in cerebellar diffuse plaques: enzyme-linked immunosorbent assay and immunocytochemical study", Brain Res. 679: 151-156 (1995).
Tamaoka et al., "Antibodies to amyloid beta protein (A beta) crossreact with glyceraldehydes-3-phosphate dehydrogenase (GAPDH)," Neurobiology of Aging 17(3):405-414 (1996).
Tamaoka et al., "Biochemical Evidence for the Long-Tail Form (Aβ1-42/43) of Amyloid β Protein as a Seed Molecule in Cerebral Deposits of Alzheimer's Disease", Biochem. Biophys. Res. Comm. 205(1): 834-842 (1994).
Tanzi et al., "Amyloid β Protein Gene: cDNA, mRNA Distribution, and Genetic Linkage Near the Alzheimer Locus," Science, 235:880-884 (1987).
Tanzi et al., "Protease inhibitor domain encoded by an amyloid protein precursor mRNA associated with Alzheimer's disease," Nature, 331:528-530 (1988).
Tomita et al., "The presenilin 2 mutation (N141I) linked to familial Alzheimer disease (Volga German families) increases the secretion of amyloid β protein ending at the 42nd (or 43rd) residue," PNAS, 94:2025-2030 (1997).
Travis, J., "New Piece in Alzheimer's Puzzle," Science, 261:828-829 (1993).
Tsuzuki et al., "Amyloid β protein in rat soleus in choroquine-induced myopthy using end-specific antibodies for Aβ40 and Aβ42: immunohistochemical evidence for amyloid β protein," Neuroscience Letters, 2002:77-80 (1995).
Turner et al., "Amyloids β40 and β42 Are Generated Intracellularly in Cultured Human Neurons and Their Secretion Increases with Maturation", J. Biol. Chem. 271(15): 8966-8970 (1996).
U.S. Appl. No. 08/665,649 Advisory Action mailed Mar. 29, 2000.
U.S. Appl. No. 08/665,649 Advisory Action US mailed Dec. 2, 1998.
U.S. Appl. No. 08/665,649 Final Office Action mailed Jul. 1, 2002.
U.S. Appl. No. 08/665,649 Final Office Action mailed May 27, 1998.
U.S. Appl. No. 08/665,649 Final Office Action mailed Nov. 4, 1999.
U.S. Appl. No. 08/665,649 First Non-Final Office Action mailed Aug. 19, 2997.
U.S. Appl. No. 08/665,649 Non-Final Office Action mailed Jul. 7, 2000.
U.S. Appl. No. 08/665,649 Notice of Allowance mailed Feb. 6, 2003.
U.S. Appl. No. 08/665,649 Restriction Requirement mailed Apr. 7, 1997.
Ueda et al., "Alz-50 recognizes a phosphorylated epitope of tau protein," J. Neuroscience, 10:3295-3304 (1990).
Uno et al., "The age of biosenescence and the incidence of cerebral beta-amyloidosis in aged captive rhesus monkeys " Ann. N.Y. Acad. Sci., 695:232-235 (1993).
Usami et al., "The Triplet of Lysine Residues (Lys724-Lys725-Lys726) of Alzheimer's Amyloid Precursor protein Plays an Important Role in Membrane Anchorage and Processing," J. Neurochem., 61(1):239-246 (1993).
Van Duijn et al., "Genetic transmission of Alzheimer's disease among families in a Dutch population based study," J. Med. Genet., 30:640-646 (1993).
Van Gool et al., "Cerebrospinal Fluid Markers of Alzheimer's Disease " JAGS, 39:1025-1039 (1991).
Vigo-Pelfrey et al., "Characterization of beta-amyloid peptide from human cerebrospinal fluid," J. Neurochem. 61(5):1965-1968 (1993).
Wang et al., "Tissue- and Development-specific Expression of the Human Phenylalanine Hydroxylase/Chloramphenicol Acetyltransferase Fusion Gene in Transgenic Mice," J. Biol. Chem., 267(21):15105-15110 (1992).
Weidemann et al., "Identification, biogenesis, and localization of precursors of Alzheimer's disease A4 amyloid protein," Cell, 57:115-126 (1989).
Wells et al., "Human dystrophin expression corrects the myopathic phenotype in transgenic mdx mice," Human Molecular Genetics, 1(1):35-40 (1992).
Westphal, H., "Mouse models of human diseases," Curr. Opin. Biotech., 2:830-833 (1991).
Wiedlocha et al., "Dual Mode of Signal Transduction by Externally Added Acidic Fibroblast Growth Factor," Cell, 76:1039-1051 (1994).
Winker, M. A., "Tacrine for Alzheimer's Disease. Which patient, what dose?," JAMA, 271(13):1023-1024 (1994).
Wirak et al., "Age-Associated Inclusions in Normal and Transgenic Mouse Brain," Science, 255:1443-45 (1992).
Wirak et al., "Deposits of Amyloid β Protein in the Central Nervous System of Transgenic Mice," Science, 253:323-325 (1991).
Wirak et al., "Regulatory region of human amyloid precursor protein (APP) gene promotes neuron-specific gene expression in the CNS of transgenic mice," EMBO J., 10(2):289-296 (1991).
Wisniewski et al., "Alzheimer's disease and Soluble A.beta.," Neurobiology of Aging, 15(2):143-152 (1994).
Wisniewski, Alzheimer's Disease, eds. Becker et al., p. 206.
Wolozin et al., "A neuronal antigen in the brains of Alzheimer patients," Science, 232:648-650 (1986).
Wolozin et al., "Alzheimer-Related Neuronal Protein A68: Specificity and Distribution," Annals of Neurology, 22(4):521-526 (1987).
Wong et al., "Neuritic Plaques and Cerebrovascular Amyloid in Alzheimer Disease are Antigenically Related," PNAS, 92:8729 (1985).
Yamaguchi et al., "Immunohistochemical analysis of COOH-termini of Amyloid beta protein (A β) using end-specific antisear for Aβ40 and Aβ42 in Alzheimer's disease and normal aging", Amyloid: Int. J. Exp. Clin. Invest. 2: 7-16 (1995).
Yamaguchi, "Transgenic mice for the amyloid precursor protein 695 isoform have impaired spatial memory," NeuroReport, 2(12):781-784 (1991).
Yang et al., "Monoclonal Antibody to the C-terminus of Beta-Amyloid," Neuroreport, 16(5):2117-2120 (1994).
Yanker et al., "Neurotoxicity of a Fragment of the Amyloid Precursor Associated with Alzheimer's Disease," Science, 245:417-420 (1989).

Also Published As

Publication number Publication date
EP0906577B2 (en) 2012-07-18
WO1997048983A1 (en) 1997-12-24
JP2000514178A (en) 2000-10-24
CA2258348A1 (en) 1997-12-24
AU3572797A (en) 1998-01-07
US20030148392A1 (en) 2003-08-07
US6610493B1 (en) 2003-08-26
DE69738961D1 (en) 2008-10-16
ATE407362T1 (en) 2008-09-15
CA2258348C (en) 2012-01-03
EP0906577A1 (en) 1999-04-07
JP3694033B2 (en) 2005-09-14
EP0906577B1 (en) 2008-09-03

Similar Documents

Publication Publication Date Title
US7993627B2 (en) Methods for determining whether a compound alters the amount of at least one αβ (X-41) peptide and the amount of either total αβ or at least one αβ (X-40) peptide produced by a non-human mammal
WO1997048983A9 (en) SCREENING COMPOUNDS FOR THE ABILITY TO ALTER THE PRODUCTION OF AMYLOID-β PEPTIDE (x-≥41)
US6114133A (en) Methods for aiding in the diagnosis of Alzheimer&#39;s disease by measuring amyloid-β peptide (x-≧41)
US7700309B2 (en) Methods for aiding in the diagnosis of alzheimer&#39;s disease by measuring amyloid-β peptide (X-&gt;41) and tau
EP0667959B1 (en) Methods for identifying inhibitors of the production of beta-amyloid peptide
US5837672A (en) Methods and compositions for the detection of soluble β-amyloid peptide
US6018024A (en) Methods and compositions for monitoring cellular processing of beta-amyloid precursor protein
US5593846A (en) Methods for the detection of soluble β-amyloid peptide
AU687747C (en) Methods and compositions for the detection of soluble beta-amyloid peptide
AU722044B2 (en) Methods and compositions for the detection of soluble beta-amyloid peptide
AU2008200489A1 (en) Methods and compositions for the detection of soluble beta-amyloid peptide

Legal Events

Date Code Title Description
REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20150809